Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas

  • Authors:
    • Baihui Liu
    • Ximao Cui
    • Shan Zheng
    • Kuiran Dong
    • Rui Dong
  • View Affiliations

  • Published online on: January 2, 2017     https://doi.org/10.3892/ol.2017.5558
  • Pages: 1360-1364
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA methylation has a crucial role in cancer biology and has been recognized as an activator of oncogenes and inactivator of tumor suppressor genes, both of which are mechanisms for tumorigenesis. Kallikrein‑related peptidase 4 (KLK4), has been suggested to be an oncogene in various types of cancer. The aim of the present study was to assess the DNA methylation patterns of the KLK4 gene in cancerous samples harvested from patients with hepatoblastoma (HB). KLK4 mRNA expression levels were detected using reverse transcription‑quantitative polymerase chain reaction and assessed its DNA methylation patterns using high‑throughput mass spectrometry on a matrix‑assisted laser desorption/ionization time‑of‑flight mass array. A total of 10 HB and 10 normal liver tissue samples were obtained from patients with HB. The results of the present study showed that a significantly higher level of KLK4 mRNA expression levels were detected in HB tissues, as compared with the matched controls. Furthermore, the KLK4 gene promoter region was distinctively less methylated in the HB samples compared with the controls and negatively correlated with KLK4 mRNA expression levels. These findings indicate that aberrant methylation of KLK4 may contribute to its upregulated mRNA expression in HB.

Introduction

Hepatoblastoma (HB) is the most common type of malignant liver tumor in infants and children. Although it accounts for just 0.5–1.5 cases per million children per year, the mortality rate is 35–50% in high-risk patients (1). Previous studies have suggested an association with familial adenomatous polyposis (FAP) (2), both low and high birth weights (3), and constitutional trisomy 18 (4); however its etiology remains unknown.

Currently, alphafetoprotein levels, histological analysis, tumor resectability and tumor metastasis are the only prognosis factors for HB. Therefore, novel targets for early detection and improved therapies and prognosis are required.

Epigenetic reprogramming has a crucial role in tumorigenesis; one of the key mechanisms is DNA methylation. DNA methylation, maintained by DNA methyltransferases, refers to the addition of a methyl group to the carbon 5 of the cytosine ring in CpG dinucleotides of gene promoter regions (5). DNA methylation is the only genetically programmed DNA modification process in mammals that is involved in the regulation of various biological processes, including gene transcription, X-chromosome inactivation, genomic imprinting and chromatin modification (68). It is generally recognized that DNA methylation is a form of gene closure; the higher the level of methylation, the more silent a gene is. Therefore, the hypermethylation of suppressor genes or hypomethylation of oncogenes may contribute to tumorigenesis. This hypothesis has been investigated and supported in various studies on tumors (9), including HB (10).

Kallikrein-related peptidase 4 (KLK4), which is a member of the KLK family, is considered to be an oncogene associated with various types of cancer, including colorectal adenocarcinoma and prostate cancer (11,12). In a pilot study, we detected aberrant expression of KLK4 in HB and normal tissues, and theorized if alterations in methylation status affect its expression. Therefore, the present study aimed to investigate KLK4 methylation in 10 HB samples in order to profile genes that are differentially methylated in this disease.

Materials and methods

Patients and sample collection

The present study utilized 10 HB and adjacent non-tumor tissues from patients who underwent partial hepatectomy at the Children's Hospital of Fudan University (Shanghai, China). The patients ranged in age from 7 to 30 months. Informed consent was obtained retrospectively from clinical files and the diagnoses were confirmed by the Department of Pathology following the presence of >80% viable tumor cells (13). Clinical and pathological data are listed in Table I. The Ethics Committee of the Children's Hospital of Fudan University approved the use of these human samples. Genomic DNA was extracted according to standard procedures from 10 matched HB tumors and non-tumor tissue sections that were stored at −80°C immediately after surgery.

Table I.

Clinical and pathological characteristics of the study subjects.

Table I.

Clinical and pathological characteristics of the study subjects.

SubjectAge (months)aGenderDiagnosis typeAlpha-fetoprotein (ng/ml)
  1  7MaleMixed embryonal/fetal subtype68490
  223MaleMixed embryonal/fetal subtype>121,000
  311FemaleMixed embryonal/fetal subtype>121,000
  410FemaleMixed embryonal/fetal subtype>121,000
  520FemaleMixed embryonal/fetal subtype>121,000
  6  7MaleMixed embryonal/fetal subtype>121,000
  730MaleEpithelial type>121,000
  814MaleEpithelial type>121,000
  919MaleEpithelial type>121,000
107FemaleEpithelial type>121,000

a On the date of serum sample collection.

DNA/RNA extraction and reverse transcription quantitative-polymerase chain reaction (RT-qPCR)

DNA was extracted from 10 HB primary tumors and adjacent non-tumor tissues. DNA samples were stored at −80°C until subsequent use for mass spectrometry analysis. Total RNA was extracted from 10 HB tumor and non-tumor pairs using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), and, reverse-transcribed into cDNA using a PrimeScript RT reagent kit (Perfect Real Time) with gDNA Eraser (Takara Biotechnology Co., Ltd., Dalian, China) following the manufacturer's protocol. This was then subjected to qPCR analysis to assess the levels of KLK4 mRNA expression using a SYBRGreen PCR kit (Takara Biotechnology Co., Ltd.). The KLK4 primers used for qPCR were: Forward, 5′-GGACTCCTGCAACGGTGACTCT-3′ and reverse, 5′-TAGACACCTGGCACGCCAACTT-3′. The PCR cycling conditions were as follows: 95°C for 30 sec, followed by 95°C for 5 sec and 60°C for 34 sec, for 45 cycles. For quantitative results, KLK4 expression was represented as fold-change by the 2−ΔΔCq method and statistically analyzed (14).

Primer design and PCR tagging for EpiTYPER assay

Primers for the KLK4 gene were designed to cover the regions with the most CpG sites (Fig. 1). Selected amplicons were 500 bp upstream of the promoter of the KLK4 gene. Genomic DNA was bisulfite-treated, and its reverse primer was supplemented with a T7-promoter tag for subsequent PCR amplification.

Mass spectrometry

Following treatment with shrimp alkaline phosphatase (SAP) in vitro transcription and uracil-specific cleavage, the DNA samples were robotically dispensed onto silicon matrix preloaded chips (SpectroCHIP; Sequenom, San Diego, CA, USA). Mass spectra were collected using a MassARRAY Compact matrix-assisted laser desorption/ionization time-of-flight system (Sequenom), and the methylation ratios of the spectra were generated using EpiTYPER software v1.0 (Sequenom).

Statistical methods

Statistical analyses and graphical depiction of data were generated using GraphPad Prism 5.0 (GraphPad Software, Inc., La Jolla, CA, USA). Results are presented as the mean ± standard error of the mean and were evaluated using a two-tailed Student's t-test unless otherwise specified, in which instance unpaired t-test or Pearson's correlation analysis were used. Statistical analyses were also performed using SPSS 19.0 software (IBM SPSS, Armonk, NY, USA) for Windows. P<0.05 was considered statistically significant to indicate a statistically significant difference.

Results

KLK4 mRNA expression levels are elevated in HB

Expression levels of KLK4 mRNA were analyzed in the 10 pairs of tumor (n=10) and adjacent normal liver (n=10) tissues using RT-qPCR. Expression levels of KLK4 in HB tissues were significantly increased, as compared with matched non-tumor liver tissues (0.039±0.0077 vs. 0.004±0.0005, respectively; P<0.01; Fig. 2).

DNA methylation of KLK4 is reduced in HB

Methylation patterns of KLK4 were verified in the 10 HB samples and match non-tumor controls. Enriched methylation positions located at 500 bp upstream of the transcription initiation site were analyzed; 11/22 sites in the amplicons were amenable to analysis. Using two-way hierarchical cluster analysis, two sites were found to be at a significantly lower degree of methylation in the HB tissues, as compared with their non-tumor counterparts (Fig. 3). Furthermore, these sites exhibited a significant difference in the mean methylation levels of the KLK4 gene between HB and adjacent non-tumor tissues (0.3755±0.03218 vs. 0.4885±0.01176, respectively; P<0.01; Fig. 4).

Correlation between hypomethylation with KLK4 and expression of mRNA

Using linear Pearson's R correlation, the correlations between KLK4 mRNA expression and DNA methylation status of CpG4 and CpG9 sites harvested from the 10 samples were analyzed. This analysis demonstrated that the expression of KLK4 is negatively correlated with its methylation status (r=−0.47; P=0.037; Fig. 5).

Discussion

Pediatric solid tumors develop after relatively short latency periods, and thus are becoming one of the most common reasons for child mortality (15,16). HB is the most common liver tumor found in children, and its incidence is increasing in North America and Europe (17). Although its pathogenesis and progression have been extensively studied for the last two decades, its etiology remains to be fully elucidated. Through a previous study that involved sequencing of the HB exome, we recently detected a novel oncogene (caprin family member 2) and three tumor suppressors (speckle-type POZ protein, olfactory receptor family 5 subfamily I member 1 and cell division cycle 20B) that influence HB cell growth (18). The present group has also detected an association between long non-coding RNA (lncRNA) and HB in a previous study (19).

Hypermethylation or hypomethylation of gene promoter regions is recognized as one of the mechanisms that can silence or activate the oncogenes (20). It has recently been indicated that aberrant promoter methylation of the RAS association domain family protein 1 may contribute to the pathogenesis of HB and is considered to be a significant prognostic indicator in HB (1,21).

KLK4 is a member of the KLK family. It has previously been reported that KLK4 is overexpressed in endometrial carcinoma (22), colorectal adenocarcinoma, and prostate cancer; however, the mechanism remains unclear. Detection of KLK4 mRNA expression levels in HB and normal liver tissues by qPCR in the present study demonstrated a significant difference between tissue types. KLK4 mRNA expression levels in HB tissues were significantly higher than in normal liver tissues. In an independent cohort of 10 adjacent HB-non-tumor tissues pairs, we theorized whether there was a correlation between KLK4 mRNA expression and DNA methylation. To further understand the association between HB and KLK4 methylation, the methylation status of KLK4 genes were analyzed in the 10 paired samples. Following methylation quantification of the KLK4 gene using MALDI-TOF MS, three CpG sites were detected per sample. Hypermethylation degrees of two CpG sites in the promoter regions of the KLK4 gene for normal liver tissues were significantly higher than those of HB samples. Furthermore, correlation analysis indicated that expression of KLK4 mRNA is significantly negatively correlated with its methylation status. This finding supports previously published data, which suggested that KLK4 may be an oncogene (2326).

To the best of our knowledge, the present study is the first to examine the methylation of KLK4 genes in HB, and an association between the level of KLK4 mRNA and methylation was successfully detected. This finding is consistent with the results of previous studies, demonstrating that hypomethylation of promoter regions of crucial genes is able to activate relevant gene expression and may contribute to tumorigenesis (2729). A limitation of the present study is that only 10 pairs of patient samples were used. Additional samples are required for investigation in order to more accurately represent the population.

In conclusion, the present findings indicate that aberrant promoter methylation of KLK4 may contribute to the tumorigenesis of HB, and that KLK4 may be a potential biomarker for HB. However, since HB is an uncommon disease, its etiology, pathophysiology, and molecular mechanisms remain unknown. Further studies are required in order to fully understand and treat this disease.

Acknowledgements

This study would like to thank the following for providing financial support: The National Key Clinical Specialty Construction Programs of China (2014–2016), the Shanghai ‘Non key-in-key discipline’ Clinical medical centers (2014–2016), the Shanghai Rising-Star Program (A type) (grant no. 15QA1400800), the National Natural Science Foundation of China (grant no. 81572324) and the Science Foundation of Shanghai (grant no. 15ZR1404200).

References

1 

Honda S, Miyagi H, Suzuki H, Minato M, Haruta M, Kaneko Y, Hatanaka KC, Hiyama E, Kamijo T, Okada T and Taketomi A: RASSF1A methylation indicates a poor prognosis in hepatoblastoma patients. Pediatr Surg Int. 29:1147–1152. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Kingston JE, Herbert A, Draper GJ and Mann JR: Association between hepatoblastoma and polyposis coli. Arch Dis Child. 58:959–962. 1983. View Article : Google Scholar : PubMed/NCBI

3 

Ross JA: Hepatoblastoma and birth weight: Too little, too big, or just right? J Pediatr. 130:516–517. 1997.PubMed/NCBI

4 

Mamlok V, Nichols M, Lockhart L and Mamlok R: Trisomy 18 and hepatoblastoma. Am J Med Genet. 33:125–126. 1989. View Article : Google Scholar : PubMed/NCBI

5 

Herman JG and Baylin SB: Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 349:2042–2054. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Geiman TM and Robertson KD: Chromatin remodeling, histone modifications, and DNA methylation-how does it all fit together? J Cell Biochem. 87:117–125. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Reik W and Walter J: Genomic imprinting: Parental influence on the genome. Nat Rev Genet. 2:21–32. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Chow J and Heard E: X inactivation and the complexities of silencing a sex chromosome. Curr Opin Cell Biol. 21:359–366. 2009. View Article : Google Scholar : PubMed/NCBI

9 

O'Sullivan E and Goggins M: DNA methylation analysis in human cancer. Methods Mol Biol. 980:131–156. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Esteller M: Epigenetics in cancer. N Engl J Med. 358:1148–1159. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Kontos CK, Chantzis D, Papadopoulos IN and Scorilas A: Kallikrein-related peptidase 4 (KLK4) mRNA predicts short-term relapse in colorectal adenocarcinoma patients. Cancer Lett. 330:106–112. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Lose F, Srinivasan S, O'Mara T, Marquart L, Chambers S, Gardiner RA, Aitken JF; Australian Prostate Cancer BioResource; Spurdle AB, Batra J and Clements JA: Genetic association of the KLK4 locus with risk of prostate cancer. PloS One. 7:e445202012. View Article : Google Scholar : PubMed/NCBI

13 

Haas JE, Feusner JH and Finegold MJ: Small cell undifferentiated histology in hepatoblastoma may be unfavorable. Cancer. 92:3130–3134. 2001. View Article : Google Scholar : PubMed/NCBI

14 

Johnson MR, Wang KS, Smith JB, Heslin MJ and Diasio RB: Quantitation of dihydropyrimidine dehydrogenase expression by real-time reverse transcription polymerase chain reaction. Anal Biochem. 278:175–184. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Otte JB: Progress in the surgical treatment of malignant liver tumors in children. Cancer Treat Rev. 36:360–371. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Tiao GM, Bobey N, Allen S, Nieves N, Alonso M, Bucuvalas J, Wells R and Ryckman F: The current management of hepatoblastoma: A combination of chemotherapy, conventional resection, and liver transplantation. J Pediatr. 146:204–211. 2005. View Article : Google Scholar : PubMed/NCBI

17 

von Schweinitz D: Hepatoblastoma: Recent developments in research and treatment. Semin Pediatr Surg. 21:21–30. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Jia D, Dong R, Jing Y, Xu D, Wang Q, Chen L, Li Q, Huang Y, Zhang Y, Zhang Z, et al: Exome sequencing of hepatoblastoma reveals novel mutations and cancer genes in the Wnt pathway and ubiquitin ligase complex. Hepatology. 60:1686–1696. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Dong R, Jia D, Xue P, Cui X, Li K, Zheng S, He X and Dong K: Genome-wide analysis of long noncoding RNA (lncRNA) expression in hepatoblastoma tissues. PloS One. 9:e855992014. View Article : Google Scholar : PubMed/NCBI

20 

Baylin SB and Ohm JE: Epigenetic gene silencing in cancer-A mechanism for early oncogenic pathway addiction? Nat Rev Cancer. 6:107–116. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Harada K, Toyooka S, Maitra A, Maruyama R, Toyooka KO, Timmons CF, Tomlinson GE, Mastrangelo D, Hay RJ, Minna JD and Gazdar AF: Aberrant promoter methylation and silencing of the RASSF1A gene in pediatric tumors and cell lines. Oncogene. 21:4345–4349. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Zhang SQ, Cai B, Liu L, He YY, Yang YX and Wan XP: Kallikrein 4 overexpression in endometrial carcinoma and upregulation by estrogen via mitogen-activated protein kinase signal pathway. Int J Gynecol Cancer. 19:1377–1383. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Klokk TI, Kilander A, Xi ZJ, Waehre H, Risberg B, Danielsen HE and Saatcioglu F: Kallikrein 4 is a proliferative factor that is overexpressed in prostate cancer. Cancer Res. 67:5221–5230. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Xi ZJ, Kaern J, Davidson B, Klokk TI, Risberg B, Tropé C and Saatcioglu F: Kallikrein 4 is associated with paclitaxel resistance in ovarian cancer. Gynecol Oncol. 94:80–85. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Papachristopoulou G, Avgeris M and Scorilas A: Expression analysis and study of KLK4 in benign and malignant breast tumours. Thromb Haemost. 101:381–387. 2009.PubMed/NCBI

26 

Mangé A, Desmetz C, Berthes ML, Maudelonde T and Solassol J: Specific increase of human kallikrein 4 mRNA and protein levels in breast cancer stromal cells. Biochem Biophys Res Commun. 375:107–112. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, Herb B, Ladd-Acosta C, Rho J, Loewer S, et al: Differential methylation of tissue-and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet. 41:1350–1353. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, Cui H, Gabo K, Rongione M, Webster M, et al: The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 41:178–186. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Ogoshi K, Hashimoto SI, Nakatani Y, Qu W, Oshima K, Tokunaga K, Sugano S, Hattori M, Morishita S and Matsushima K: Genome-wide profiling of DNA methylation in human cancer cells. Genomics. 98:280–287. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 13 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu B, Cui X, Zheng S, Dong K and Dong R: Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas. Oncol Lett 13: 1360-1364, 2017
APA
Liu, B., Cui, X., Zheng, S., Dong, K., & Dong, R. (2017). Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas. Oncology Letters, 13, 1360-1364. https://doi.org/10.3892/ol.2017.5558
MLA
Liu, B., Cui, X., Zheng, S., Dong, K., Dong, R."Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas". Oncology Letters 13.3 (2017): 1360-1364.
Chicago
Liu, B., Cui, X., Zheng, S., Dong, K., Dong, R."Aberrant KLK4 gene promoter hypomethylation in pediatric hepatoblastomas". Oncology Letters 13, no. 3 (2017): 1360-1364. https://doi.org/10.3892/ol.2017.5558