Skip to main content
Erschienen in: BMC Cancer 1/2011

Open Access 01.12.2011 | Research article

Decreased expression of 17β-hydroxysteroid dehydrogenase type 1 is associated with DNA hypermethylation in colorectal cancer located in the proximal colon

verfasst von: Agnieszka Anna Rawłuszko, Karolina Horbacka, Piotr Krokowicz, Paweł Piotr Jagodziński

Erschienen in: BMC Cancer | Ausgabe 1/2011

Abstract

Background

The importance of 17β-estradiol (E2) in the prevention of large bowel tumorigenesis has been shown in many epidemiological studies. Extragonadal E2 may form by the aromatase pathway from androstenedione or the sulfatase pathway from estrone (E1) sulfate followed by E1 reduction to E2 by 17-β-hydroxysteroid dehydrogenase (HSD17B1), so HSD17B1 gene expression may play an important role in the production of E2 in peripheral tissue, including the colon.

Methods

HSD17B1 expression was analyzed in colorectal cancer cell lines (HT29, SW707) and primary colonic adenocarcinoma tissues collected from fifty two patients who underwent radical colon surgical resection. Histopathologically unchanged colonic mucosa located at least 10-20 cm away from the cancerous lesions was obtained from the same patients. Expression level of HSD17B1 using quantitative PCR and western blot were evaluated. DNA methylation level in the 5' flanking region of HSD17B1 CpG rich region was assessed using bisulfite DNA sequencing and HRM analysis. The influence of DNA methylation on HSD17B1 expression was further evaluated by ChIP analysis in HT29 and SW707 cell lines. The conversion of estrone (E1) in to E2 was determined by electrochemiluminescence method.

Results

We found a significant decrease in HSD17B1 transcript (p = 0.0016) and protein (p = 0.0028) levels in colorectal cancer (CRC) from the proximal but not distal colon and rectum. This reduced HSD17B1 expression was associated with significantly increased DNA methylation (p = 0.003) in the CpG rich region located in the 5' flanking sequence of the HSD17B1 gene in CRC in the proximal but not distal colon and rectum. We also showed that 5-dAzaC induced demethylation of the 5' flanking region of HSD17B1, leading to increased occupation of the promoter by Polymerase II, and increased transcript and protein levels in HT29 and SW707 CRC cells, which contributed to the increase in E2 formation.

Conclusions

Our results showed that reduced HSD17B1 expression can be associated with DNA methylation in the 5' flanking region of HSD17B1 in CRC from the proximal colon.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-11-522) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AAR contributed to designing the study, acquisition of data, analysis and interpretation of data, and in writing the manuscript. KH participated in the acquisition and interpretation of the data. As Principal Investigators, PK and JPP were involved in the intellectual and experimental programming of the study, the interpretation of data, and writing the manuscript. All authors read and approved the final manuscript.

Background

Colorectal cancer (CRC) is the third in the United States [1] and second in Europe [2] cause of malignant disease deaths among adults. Population based studies have shown significant gender differences in CRC incidence and mortality [3]. CRC occurrence is more frequent among men than in pre-menopausal women, which suggests a protective role of 17-β-estradiol (E2) in the development of this cancer [35]. Moreover, many case-control and cohort studies showed an inverse relationship between the risk of CRC incidence and the use of hormone replacement therapy by post-menopausal women [6, 7]. Although E2 is mainly biosynthesized by the ovaries, this hormone can also be produced in peripheral tissues in both genders [8]. Extragonadal E2 can be formed by the aromatase pathway from the androstenedione or the sulfatase pathway from estrone (E1) sulfate, followed by E1 reduction to E2 by 17-β-hydroxysteroid dehydrogenase (HSD17B1) [8, 9]. Therefore, HSD17B1 gene expression may play an important role in the production of E2 in peripheral tissue. HSD17B1 transcription may start from distal or proximal promoters, forming, respectively, the long (2.3-kb) or short (1.3-kb) transcript, but the short mRNA is thought to be translated into the HSD17B1 protein (Additional file 1) [1013].
Epigenetics involve variable usage of DNA due to chromatin modifications without disturbances in the DNA sequence [14]. The low level of DNA methylation within 5'-CpG-3' dinucleotides may relate to the activation of transcription or chromosomal instability [14, 15]. It has been shown that, similarly to genetic mutations, hypermethylation or hypomethylation of gene promoters may change the expression of cancer related genes in different malignancies, including CRC [14]. DNA methylation is carried out by DNA methyltransferases (DNMTs), and increased levels of some DNMTs account for transcriptional silencing of cancer protective genes [15].
We studied HSD17B1 transcript and protein levels in primary cancerous tissue and histopathologically unchanged colorectal tissue from the same fifty two patients with CRC. We also evaluated the effect of 5-Aza-2'-deoxycytidine (5-dAzaC), a DNMTs inhibitor, on HSD17B1 transcript and protein levels in HT29 and SW707 CRC cells. Moreover, we determined the level of methylation in the 5' flanking region of HSD17B1 in primary cancerous and histopathologically unchanged colonic tissues as well as HT29 and SW707 CRC cells treated with 5-dAzaC.

Methods

Patient material

Primary colonic adenocarcinoma tissues were collected between June 2009 and June 2010 from fifty two patients who underwent radical colon surgical resection at the Department of General and Colorectal Surgery, Poznań University of Medical Sciences, Poland (Table 1). Histopathologically unchanged colonic mucosa located at least 10-20 cm away from the cancerous lesions was obtained from the same patients. Samples were immediately snap-frozen in liquid nitrogen and stored at −80 º C until RNA/DNA/protein isolation. At the time of surgery, the median and mean age of the patients was 70 years (range 39-85) and 68.36 ± 11.7 years, respectively. Two of the patients exhibited T1 tumour stage, nine of the patients exhibited T2 tumour stage, thirty seven patients exhibited T3 tumour stage and four patients exhibited T4 tumour stage. Written informed consent was obtained from all participating individuals. The procedures of the study were approved by the Local Ethical Committee of Poznań University of Medical Sciences.
Table 1
HSD17B1 transcript and protein levels in primary cancerous and histopathologically unchanged tissue samples from patient with CRC.
n = 52
Primary cancerous tissues
Histopathologically unchanged tissues
Pc
  
Median (range)
Mean (± SD)
Median (range)
Mean (± SD)
 
Age (Years)
      
<60
10
3.511 (2.109-3.868) a
3.282 ± 0.5921 a
3.415 (2.726-5.107)a
3.611 ± 0.6309 a
0.2450a
  
2.634 (1.681-3.218)b
2.579 ± 0.5243b
2.620 (2.108-3.398)b
2.719 ± 0.4843b
0.5423b
>60
42
3.531 (2.071-5.227) a
3.569 ± 0.6649 a
3.692 (2.790-5.785)a
3.834 ± 0.6251a
0.1087a
  
2.728 (1.728-4.023)b
2.720 ± 0.4702b
2.952 (1.364-3.472)b
2.835 ± 0.4913b
0.1121b
Gender
      
Female
20
3.384 (2.109-4.455) a
3.371 ± 0.5676 a
3.449 (2.726-5.785)a
3.719 ± 0.7333a
0.2340 a
  
2.793 (1.681-3.218)b
2.666 ± 0.4767b
2.811 (2.108-3.472)b
2.844 ± 0.4065b
0.2119b
Male
32
3.571 (2.071-4.653) a
3.557 ± 0.6523 a
3.857(2.790-5.259) a
3.890 ± 0.5855 a
0.0388 a
  
2.702 (1.728-4.023)b
2.713 ± 0.4941b
2.962 (1.364-3.449)b
2.792 ± 0.5395b
0.2832b
CRC localization
      
Proximal colon (cecum to transverse)
20
3.507(2.666-4.455) a
3.427 ± 0.4350 a
3.874 (3.050-5.785) a
4.057 ± 0.7488 a
0.0016 a
  
2.793 (1.681-3.218)b
2.666 ± 0.4767b
2.811 (2.108-3.472)b
2.844 ± 0.4065b
0.0028b
Distal colon (splenic flexture to sigmoid)
12
3.214 (2.071-4.313) a
3.218 ± 0.7760 a
3.449 (2.726-4.587) a
3.630 ± 0.6372 a
0.1685 a
  
2.798 (1.772-3.254)b
2.691 ± 0.4777b
2.860 (1.364-3.391)b
2.619 ± 0.6350b
0.7763b
Rectum
20
3.544 (2.523-4.653) a
3.657 ± 0.6011 a
3.564 (3.226-4.862) a
3.762 ± 0.500 a
0.8839 a
  
2.951 (1.728-3.398)b
2.815 ± 0.4436b
2.988 (2.019-3.383)b
2.913 ± 0.3871b
0.5019b
Histologic grade
      
G1
4
4.059 (2.362-4.402) a
3.721 ± 0.9467 a
4.431 (3.123-5.259) a
4.311 ± 0.9076 a
0.4026 a
  
2.926 (2.586-3.040)b
2.870 ± 0.2060b
2.789 (2.026-3.147)b
2.688 ± 0.5370b
0.5506b
G2
35
3.379 (2.109-4.621) a
3.363 ± 0.5385 a
3.524 (2.726-5.785) a
3.681 ± 0.5619 a
0.0335 a
  
2.647 (1.681-4.023)b
2.649 ± 0.4993b
2.933 (2.019-3.472)b
2.863 ± 0.4114b
0.0659b
G3
13
3.875 (2.071-4.839) a
3.807 ± 0.7092 a
4.043 (2.790-5.227) a
4.165 ± 0.7346 a
0.2587 a
  
2.676 (1.845-3.431)b
2.670 ± 0.5098b
3.023 (1.364-3.449)b
2.763 ± 0.6858b
0.5114b
The HSD17B1 transcript levelsa were standardized by the geometric mean of PBGD and hMRPL19 cDNA and expressed as decimal logarithm of multiples of these cDNA copies in calibrator. b The amount of western blot-detected proteins was presented as the decimal logarithm HSD17B1 to β-actin band optical density ratio. c Unpaired, two-tailed t-test or U-Mann-Whitney test.

Antibodies and Reagents

Goat polyclonal (Gp) anti-HSD17B1 antibodies (Ab) (C-18), donkey anti-goat horseradish peroxidase (HRP)-conjugated Ab, anti-actin HRP-conjugated Ab (clone I-19) and anti-Polymerase II (Pol II)-rabbit polyclonal Ab (H-224) were provided by Santa Cruz Biotechnology (Santa Cruz, CA). E1, 5-dAzaC was purchased from Sigma-Aldrich Co. (St. Louis, MO). Goat anti-rabbit HRP conjugated Ab was provided by DAKO (Glostrup, Denmark).

Cell culture

HT29 colon cancer cells were obtained from the American Type Culture Collection (Rockville, MD) and SW707 CRC cells were kindly provided by the Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences. These cells were cultured in DMEM GibcoBRL (Grand Island, NY) containing 10% heat-inactivated fetal bovine serum (FBS) and 2 mM glutamine. To determine the effect of 5-dAzaC on HSD17B1 transcript and protein levels, the HT29 and SW707 cells were cultured for 24 h in phenol red-free DMEM GibcoBRL (Grand Island, NY) supplemented with 10% charcoal-dextran-stripped FBS from Sigma-Aldrich Co. (St. Louis, MO). Cells were then cultured for 6, 12, 24, 36 and 48 h either in the absence or in the presence of 5-dAzaC at concentrations of 0.33, 0.66 and 1.00 μM. These cells were used for total RNA isolation, western blotting, chromatin immunoprecipitation (ChIP) assay, and bisulfite sequencing.

Reverse transcription and real-time quantitative polymerase chain reaction (RQ-PCR) analysis of the HSD17B1 transcript levels

The total RNA from primary tissues of patients with CRC and from HT29 and SW707 cells was isolated according to the method of Chomczyński and Sacchi (1987) [16]. RNA samples were treated with DNase I, quantified, and reverse-transcribed into cDNA. RQ-PCR was carried out in a Light Cycler real-time PCR detection system from Roche Diagnostics GmbH, (Mannheim, Germany) using SYBR® Green I as detection dye. The target cDNA was quantified by relative quantification method using a calibrator for primary tissue or respective controls for HT29 and SW707 cells. The calibrator was prepared as a cDNA mix from all of the patients' samples and successive dilutions were used to create a standard curve as described in Relative Quantification Manual Roche Diagnostics GmbH, (Mannheim, Germany). For amplification, 1 μl of cDNA solution was added to 9 μl of IQ™ SYBR® Green Supermix Bio-Rad Laboratories Inc. (Hercules, CA) and primers (Additional file 1, Additional file 2). The quantity of transcript of HSD17B1 in each sample was standardized by the geometric mean of porphobilinogen deaminase (PBGD) and human mitochondrial ribosomal protein L19 (hMRPL19) cDNA levels (Additional file 2) [17, 18]. The transcript levels in patient tissues were expressed as multiplicity of these cDNA concentrations in the calibrator. In HT29 and SW707 cells, transcript levels were presented as multiplicity of the respective controls.

Western blotting analysis

Primary tissue from patients with CRC and HT29 and SW707 cells were treated with lysis RIPA buffer, and 40 μg of protein were resuspended in sample buffer and separated on 12% Tris-glycine gel using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Gel proteins were transferred to a polyvinylidene fluoride (PVDF) membrane, which was blocked with 5% milk in Tris/HCl saline/Tween buffer. Immunodetection of bands was performed with Gp anti-HSD17B1 Ab (C-18), followed by incubation with donkey anti-goat HRP-conjugated Ab. To ensure equal protein loading of the lanes, the membrane was re-stripped and incubated with anti-actin HRP-conjugated Ab (clone I-19). Bands were revealed using ECL kit and Hyperfilm ECL Amersham (Piscataway, NJ). The amounts of HSD17B1 protein was presented as the HSD17B1 to-β-actin band optical density ratio. For HT29 and SW707 cells cultured in the absence of 5-dAzaC, the ratio of HSD17B1 to β-actin was assumed to be 1.

DNA methylation evaluation by bisulfite sequencing

Genomic DNA was isolated using DNA Mammalian Genomic Purification Kit purchased from Sigma-Aldrich Co. (St. Louis, MO). One μg of genomic DNA was subjected to bisulfite conversion of cytosine to uracil according to EZ DNA Methylation Kit™ procedure from Zymo Research Corporation (Orange, CA). The position of CpG islands (chr17: 37 953 426 - 37 954 646) and binding sites of transcription factors located in the 5' flanking region of the promoter was determined by online programs [1921] (Additional file 1). A DNA fragment containing 31 CpG dinucleotides was amplified from the bisulfite-modified DNA by the primer pair (chr17: 37 953 392-37 953 917) (Additional file 1, Additional file 2) complementary to the bisulfite-DNA modified sequence. PCR amplification was performed by FastStart Taq DNA Polymerase from Roche Diagnostic GmbH (Penzberg, Germany). The PCR products were purified using Agarose Gel DNA Extraction Kit Roche (Mannheim, Germany) with subsequent cloning into pGEM-T Easy Vector System I Promega (Madison, WI) and transformation into TOPO10 E. coli strain cells. Plasmid DNA isolated from ten positive bacterial clones was used for commercial sequencing of the cloned fragment of DNA. The results of bisulfite sequencing were assessed and presented using BiQ analyzer software and the Bisulfite sequencing Data Presentation and Compilation (BDPC) web server, respectively [22, 23].

DNA methylation assessment by high resolution melting (HRM) analysis

Methylation levels of three DNA fragments located within the CpG rich region (Additional file 1) were determined by Real Time PCR amplification of bisulfite treated DNA, followed by HRM profile analysis by CFX96™ Real-Time PCR Detection System, Bio-Rad Laboratories Inc. (Hercules, CA). For PCR amplification, 1 μl solution of bisulfite treated DNA, from patients or standards, and primers (Additional file 1, Additional file 2) was added to 19 μl of 5 × Hot FIREPol EvaGreen HRM Mix, Solis BioDyne Co. (Tartu, Estonia). Standards of DNA methylation percentage were prepared by mixing in different ratio methylated and non-methylated bisulfite treated DNA from Human Methylated/Non-methylated DNA Set Zymo Research Corp. (Orange, CA). To determine the percentage of methylation, the HRM profiles of patient DNA PCR products were compared with HRM profiles of standard DNA PCR product [24] (Additional file 3). HRM methylation analysis was performed using Precision Melt Analysis™ Software, Bio-Rad Laboratories Inc. (Hercules, CA). Each PCR amplification and HRM profile analysis was performed in triplicate. The methylation for each patient was presented as a percentage of methylation in three amplified fragments located in the CpG rich region of HSD17B1.

ChIP analysis

HT29 and SW707 cells were maintained in phenol red-free DMEM GibcoBRL supplemented with 10% charcoal-dextran-stripped FBS for 6, 12, 24, 36 and 48 h either in the absence or in the presence of 5-dAzaC at a concentration of 1.00 μM. Cells were then fixed by the addition of 270 μL of 37% formaldehyde to 10 mL of culture medium for 10 min at 37 ºC, and harvested. Chromatin from 106 cells was sheared by sonicator and precleared with salmon sperm DNA-saturated protein G sepharose. The ChIP assay was conducted using Abcam ChIP Kit ab500 [25]. Chromatin was incubated with 2 μg of rabbit polyclonal anti-Pol II Ab (H-224) overnight at 4 ºC. The input and immunoprecipitated DNA were used as templates for Real Time PCR preformed using the primers corresponding to the promoter of the HSD17B1 gene (Additional file 1, Additional file 2). The percentage of HSD17B1 promoter that was bound with Pol II was calculated based on the differences between CT values for input and DNA samples from cells treated or untreated with 5-dAzaC.

Effect of 5-dAzaC on the conversion of E1 to E2 in HT29 and SW707 cells

To determine the effect of 5-dAzaC on the conversion of E1 to E2, the HT29 and SW707 cells were maintained for 24 h in phenol red-free DMEM GibcoBRL supplemented with 10% charcoal-dextran-stripped FBS from Sigma-Aldrich Co. Cells were then cultured for 36 h either in the absence or in the presence of 5-dAzaC at a concentration of 1.00 μM for HT29 and SW707 cells. These cells were then further cultured either in the absence or the presence of E1 at a concentration of 10−2 mM. After 2, 4, 6, 8, 10 and 12 h, 0.5 ml of the medium was collected and the concentration of E2 was determined by electrochemiluminescence method using Cobas 6000 Roche Diagnostics GmbH (Mannheim, Germany). The results were expressed as E2 concentration in culture medium in nM per μg cell protein.

Statistical analysis

Data groups for cell lines were assessed by ANOVA to evaluate if there was significance (P < 0.05) between the groups. For all experimental groups, which fulfilled the initial criterion, individual comparisons were performed by post hoc Tukey test with the assumption of two-tailed distribution and two samples with equal variance at the P < 0.05 level. Statistical significance was designated by asterisks in the figures.
The normality of the observed patient data distribution was assessed by Shapiro-Wilk test, and parametric unpaired, two-tailed t-test or nonparametric U-Mann-Whitney test was used to compare the mean values. P < 0.05 was considered statistically significant. The correlation between mRNA and protein levels was evaluated by Spearman nonparametric correlation coefficient method. The statistical analysis was performed with STATISTICA 6.0 software.

Results

HSD17B1 transcript and protein levels in primary cancerous and histopathologically unchanged tissues from patients with CRC

To compare HSD17B1 transcript and protein levels in cancerous and histopathologically unchanged tissues from patients with CRC we used RQ-PCR and western blotting analysis, respectively. We found significantly lower levels of HSD17B1 transcript (p = 0.0016) and protein (p = 0.0028) in primary cancerous tissues than in histopathologically unchanged tissues in patients with CRC in the proximal colon (Figure 1A,B,C and Table 1). Moreover, we observed a moderate correlation between mRNA and protein in cancerous (p = 0.0502) and histopathologically unchanged tissue (p = 0.0722) in the proximal colon. We also observed significantly lower amounts of HSD17B1 transcript (p = 0.0335, p = 0.0388) and protein (p = 0.0659, p = 0.2832) in cancerous tissues compared to histopathologically unchanged tissues in patients with G2 histological grade and male patient group, respectively. There were no significant differences in transcript (p = 0.1685, p = 0.8839) and HSD17B1 protein (p = 0.7763, p = 0.5019) levels between primary cancerous and histopathologically unchanged tissues in patients with CRC located in the distal colon and rectum (Table 1). We also did not observe differences in HSD17B1 transcript and protein levels between these tissues in the female patient group, different ages, G1/G3 histological grade (Table 1), and different tumour stage (not shown).

DNA methylation levels in primary cancerous and histopathologically unchanged tissues from patients with CRC

To compare DNA methylation levels in the HSD17B1 CpG rich region between DNA from cancerous and histopathologically unchanged tissues, we performed sodium bisulfite DNA sequencing and HRM analysis (Additional file 1, Additional file 2). Bisulfite sequencing was used for preliminary evaluation of DNA methylation changes in randomly selected patients. We observed the same pattern of methylation within all individual clones of each patient. Moreover, this evaluation showed significant differences in DNA methylation levels between cancerous and histopathologically unchanged tissues from five patients with CRC in the proximal colon (Figure 2A). However, we did not observe this difference in five patients with CRC located in the distal colon (Figure 2B) or rectum (not shown). We applied HRM analysis of PCR amplified bisulfite treated DNA to extend the DNA methylation studies in the HSD17B1 CpG rich region for cancerous and histopathologically unchanged tissues from all fifty two patients. In patients with CRC located in the proximal colon, we found a significantly higher DNA methylation percentage in cancerous than in histopathologically unchanged tissues (p = 0.003) (Table 2). However, there were no significant differences in DNA methylation percentage between cancerous and histopathologically unchanged tissues from patients with CRC located in the distal colon (p = 0.7498) and rectum (p = 0.9810) (Table 2). We also did not find significant differences in the DNA methylation percentage between these tissues in patients divided based on gender, age, histological grade (Table 2) and tumour stage (not shown).
Table 2
The percentage of DNA methylation in CpG rich regions in primary cancerous and histopathologically unchanged tissue samples from patients with CRC.
n = 52
Primary cancerous tissues
Histopathologically unchanged tissues
pa
  
Median (range)
Mean (± SD)
Median (range)
Mean (± SD)
 
Age
      
<60
10
21.05 (12.89-25.79)
20.66 ± 4.205
19.47 (13.16-28.42)
19.32 ± 4.875
0.5181
>60
42
20.00 (4.211-33.16)
20.91 ± 7.074
18.95 (2.105-36.21)
18.42 ± 6.779
0.1076
Gender
      
Female
20
24.21 (16.84-33.16)
24.42 ± 4.516
23.68 (13.68-28.42)
21.75 ± 4.499
0.1155
Male
32
18.95 (4.211-32.11)
19.51 ± 7.255
17.37 (2.105-36.21)
18.43 ± 7.206
0.5594
CRC localization
      
Proximal colon (cecum to transverse)
20
24.21 ( 13.68-33.16)
23.54 ± 5.793
17.63 ( 2.105435.79)
17.61 ± 6.050
0.0030
Distal colon (splenic flexture to sigmoid)
12
19.47 (4.211-28.95)
19.69 ± 7.159
18.95 (10.53-25.26)
18.85 ± 4.747
0.7498
Rectum
20
19.47 (6.316-28.42)
19.36 ± 6.292
19.47 (4.737-36.21)
19.30 ± 7.851
0.9810
Histologic grade
      
G1
4
21.32 (13.68-28.42)
21.18 ± 8.067
17.89 (2.105-27.89)
16.45 ± 10.97
0.5126
G2
35
20.00 (10.53-30.53)
21.17 ± 5.293
19.47 (4.737-28.42)
18.93 ± 5.884
0.0996
G3
13
21.84 (4.211-33.16)
20.21 ± 9.397
15.53 (12.63-36.21)
18.11 ± 6.727
0.4021
The primary cancerous and histopathologically unchanged tissue samples from the same patients were used for genomic DNA isolation, followed by bisulfite conversion of cytosine to uracil. The three DNA fragments of the CpG rich region were then amplified by three pairs of primers complementary to the bisulfite-DNA modified sequence (Additional file 1, Additional file 2). To determine the percentage of methylation, the HRM profiles of the patients' DNA PCR products were compared to HRM profiles of the prepared standard PCR products. DNA methylation for each patient was presented as a percentage of methylation in three DNA amplified fragments located in the CpG rich region (Additional file 1). a Unpaired, two-tailed t-test.

5-dAzaC effects on HSD17B1 transcript and protein contents in HT29 and SW707 colorectal cancer cells

We observed that 5-dAzaC resulted in a progressive increase in HSD17B1transcript levels in HT29 and SW707 cells (Figure 3A,B). For HT29 cells, we found approximately a 1.91- fold significant increase in HSD17B1 transcript levels at 48 h of incubation (Figure 3A). There was also an approximately 1.35-fold significant increase in HSD17B1 mRNA in SW707 cells at 48 h of incubation (Figure 3B). These alterations in HSD17B1 transcript levels in both HT29 and SW707 cells were associated with increased HSD17B1 protein levels (Figure 4). Densitometric analysis of western blotting bands indicated a gradual increase of HSD17B1 protein levels in a dose dependent manner at 24, 36, and 48 h of incubation for HT29 cells, and at 48 h for SW707 cells (Figure 4). Incubation of HT29 cells with 5-dAzaC at a concentration of 1.00 μM for 48 h resulted in a 2.28-fold increase in HSD17B1 protein contents (Figure 4). There was also an approximately 1.57-fold increase in HSD17B1 protein contents in SW707 cells incubated with 5-dAzaC at a concentration of 1.00 μM for 48 h as compared with the respective control (Figure 4).

5-dAzaC induced transcription of HSD17B1 via DNA demethylation in the CpG rich region and increased the binding of Pol II to the promoter in HT29 colon cancer cells

In order to assess the effect of 5-dAzaC on DNA demethylation and Pol II binding to the promoter we used bisulfite DNA sequencing and the ChIP assay, respectively (Additional file 1). We noticed the same pattern of methylation within all analysed clones. Moreover, we observed significant DNA demethylation in the CpG rich region in HT29 cells cultured for 48 h in the presence of 5-dAzaC (Figure 5A). Incubation with 5-dAzaC at a concentration of 1.00 μM for 6, 12, 24, 36, and 48 h caused an approximately 1.56 to 6.70-fold increase in the percentage of promoter occupancy by Pol II in HT29 colon cancer cells (Figure 5B). However, in SW707 CRC cells treated with 5-dAzaC at a concentration of 1.00 μM, we observed slight DNA demethylation and slight increased occupancy percentage of the promoter by Pol II (Figure 5A,B).

5-dAzaC increased the conversion of E1 to E2 in HT29 and SW707 colorectal cancer cells

5-dAzaC caused a progressive increase in the conversion of E1 to E2 in a time dependent manner in 5-dAzaC pretreated HT29 cells at 6, 8, 10 and 12 h of incubation (Figure 6A). Incubation of 5-dAzaC pretreated HT29 cells with E1 for 12 h resulted in a 3.0-fold increase in E2 levels as compared to 5-dAzaC untreated cells (Figure 6A). There was also a slight, 1.2-fold, increase in E2 concentration in 5-dAzaC pretreated SW707 cells at 12 h of incubation as compared with the respective control (Figure 6B).

Discussion

There are many environmental, dietary, lifestyle, behavioral, genetic and epigenetic factors that have been considered as risk factors for developing CRC [2630]. Malignant transformation of colonic mucosa to adenoma and adenocarcinoma is associated with the accumulation of mutations in various oncogenes, tumour suppressor genes (TSGs) and chromosome, and microsatellite instability [29, 31]. Alternatively, the epigenetic mechanism has also been considered as a strong contributor to CRC incidence. Aberrant methylation of DNA may mimic DNA mutation, leading to transcriptional silencing of TSGs and chromosome instability [15].
Accessibility of E2 is another significant factor contributing to the development of CRC. The randomized clinical trial conducted by the Women's Health Institute showed that treatment of postmenopausal women with E2 plus progestin was associated with a decreased risk of CRC [32]. In contrast, treatment of these women with E2 alone exhibited a rather slight increased risk of CRC [32]. However, the importance of E2 in the prevention of large bowel tumorigenesis has been shown in other epidemiological studies [3, 5, 33]. E2 exerts an apoptotic effect in CRC cells mainly via interaction with the estrogen receptor (ER) [3436]. This apoptotic effect of E2 in CRC can be inhibited by DNA methylation and silencing of transcription of the ESR2 gene encoding ERβ [35, 37]. Papaxoinis et al. [38] showed that ESR2 expression is higher in normal epithelium of the proximal colon as compared to the distal colon. This indicated that epigenetic silencing of ESR2 appearing in 90% of CRC cases could mainly be associated with carcinogenesis in the proximal colon [39]. Moreover, the cohort studies indicated that, in postmenopausal women, CRC more frequently developed in the proximal colon than in the distal colon [40, 41]. These findings may indicate that the proximal colon may be the major bowel region, compared to the distal colon, in which E2 may mainly exert its anticancer action [42]. Fiorelli et al. (2002) showed the involvement of HSD17B1 in the conversion of E1 to E2 in human colon cancer cells [43]. This may suggest that the extragonadal production of E2 from E1, especially in the large bowel, may be critical in CRC incidence in postmenopausal women and in men.
During CRC development, epithelial cells acquire peculiar features which enable them to grow aberrantly, ignore growth inhibition, and resist death signals [44]. We found a significant reduction of HSD17B1 transcript and protein levels in CRC in the proximal colon. However, these differences were not found in CRC in the distal colon and rectum. The decreased expression of HSD17B1 was associated with significantly increased DNA methylation in the CpG rich region located in the 5' flanking region of the HSD17B1 gene in CRC in the proximal colon but not in the distal colon and rectum.
Abnormalities in DNA methylation are observed from the early stages of lesions in aberrant crypt foci and hyperplastic polyps considered to be CRC incidence risk factors [45, 46]. To date, transcriptional silencing by DNA methylation has been demonstrated in hMLH1, PDZ and LIM domain 2, UDP-glucuronosyltransferase 1A1, HOXB13, CXCL12 and other genes in CRC [31, 4750]. Moreover, there is a distinction between tumorigenesis associated with DNA methylation and gene expression when comparing the proximal colon to the distal colon and rectum [30, 51]. Carcinogenesis in the proximal colon is mainly associated with epigenetic aberrations, whereas cancer incidence in the distal colon and rectum is mainly linked to the accumulation of genetic mutations [51]. Glebov et al. (2003) used microarray analysis to demonstrate that more than 1000 genes are expressed differently in CRC depending on its location in either the ascending or descending colon [52]. Toyota et al. (1999) characterized the CpG island methylator phenotype (CIMP) in CRC [53]. CRC with CIMP-high develops more frequently in the proximal colon, while CIMP-low or CIMP-negative is likely to be found in the distal colon [30, 51]. Hypermethylation of the promoter region of hMLH1, encoding a component of the DNA mismatch repair system, was also more frequently found in CRC in the proximal colon than in the distal colon [31].
In order to determine the role of methylation of the CpG rich region in HSD17B1 expression, we treated HT29 and SW707 cells with 5-dAzaC. We observed DNA demethylation within the CpG rich region (chr17: 37 953 426-37 954 646) (Additional file 1), which was associated with an increase in Pol II binding to the promoter and increased the HSD17B1 transcript and protein levels in HT29 cells. However, 5-dAzaC treatment caused lower DNA demethylation and binding of Pol II to promoter in SW707 cells than in HT29 cells. These differences between HT29 and SW707 cells might be due to their distinct regions of origin from the large bowel: SW707 cells were derived from the rectum, whereas HT29 originated from the colon [54, 55].
Data from the Encyclopedia of DNA elements project showed that the analyzed CpG rich region upstream of HSD17B1 (chr17: 37 953 426-37 954 646) is able to bind various transcription factors. Moreover, the enrichment of histone modification and DNAse I hypersensitivity of that region suggest promoter or enhancer activity [56]. Lornicz et al. (2004) showed that DNA methylation in intragenic and intergenic CpG rich regions may convert euchromatin to heterochromatin and lead to reduced Pol II action [57]. Based on this observation, we hypothesize that hypermethylation of the 5' flanking region of the HSD17B1 gene might inhibit the binding of transcription factors and decrease HSD17B1 expression in CRC.
5-dAzaC has been considered in the treatment of CRC. Genome-wide studies, including promoter methylation and expression microarray analysis, revealed a number of genes that are up-regulated in SW116 and Colo-320 CRC cells treated with 5-dAzaC [58]. This DNMTs inhibitor also results in DNA demethylation and induces the expression of PDZ and LIM domain 2, UDP-glucuronosyltransferase 1A1, insulin-like growth factor binding protein 7, and NGX6 genes in CRC cells [47, 48, 59, 60]. Lin et al. (2008) demonstrated that 5-dAzaC induced apoptosis and inhibited the growth, migration and invasion of colon cancer cells [60].

Conclusion

Our results showed increased DNA methylation in the 5' flanking region of HSD17B1 in CRC from the proximal colon as compared to normal colonic epithelium. These epigenetic differences were associated with a significant reduction of HSD17B1 transcript and protein levels in cancer located in the proximal colon. We also found that 5-dAzaC, an inhibitor of DNMTs, induced the demethylation of the 5' flanking region of HSD17B1, leading to increased transcript and protein levels in HT29 colon cancer cells, which contributed to the increase in E2 formation.
Although we presented that HSD17B1 expression in CRC can be epigenetically down-regulated, further studies are required to assess the concentration of endogenous E2 in normal colonic tissue and the role of endogenous E2 in the prevention of carcinogenesis.

Acknowledgements

Supported by grant No 502-01-01124182-07474, Poznań University of Medical Sciences. The authors wish to acknowledge Dr. Margarita Lianeri for her assistance.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AAR contributed to designing the study, acquisition of data, analysis and interpretation of data, and in writing the manuscript. KH participated in the acquisition and interpretation of the data. As Principal Investigators, PK and JPP were involved in the intellectual and experimental programming of the study, the interpretation of data, and writing the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Jemal A, Siegel R, Xu J, Ward E: Cancer statistics, 2010. CA Cancer J Clin. 2010, 60 (5): 277-300. 10.3322/caac.20073.CrossRefPubMed Jemal A, Siegel R, Xu J, Ward E: Cancer statistics, 2010. CA Cancer J Clin. 2010, 60 (5): 277-300. 10.3322/caac.20073.CrossRefPubMed
2.
Zurück zum Zitat Ferlay J, Parkin DM, Steliarova-Foucher E: Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 2008, 46 (4): 765-781.CrossRef Ferlay J, Parkin DM, Steliarova-Foucher E: Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 2008, 46 (4): 765-781.CrossRef
3.
Zurück zum Zitat Brenner H, Hoffmeister M, Arndt V, Haug U: Gender differences in colorectal cancer: implications for age at initiation of screening. Br J Cancer. 2007, 96 (5): 828-831. 10.1038/sj.bjc.6603628.CrossRefPubMedPubMedCentral Brenner H, Hoffmeister M, Arndt V, Haug U: Gender differences in colorectal cancer: implications for age at initiation of screening. Br J Cancer. 2007, 96 (5): 828-831. 10.1038/sj.bjc.6603628.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Ries LA, Wingo PA, Miller DS, Howe HL, Weir HK, Rosenberg HM, Vernon SW, Cronin K, Edwards BK: The annual report to the nation on the status of cancer, 1973-1997, with a special section on colorectal cancer. Cancer. 2000, 88 (10): 2398-2424. 10.1002/(SICI)1097-0142(20000515)88:10<2398::AID-CNCR26>3.0.CO;2-I.CrossRefPubMed Ries LA, Wingo PA, Miller DS, Howe HL, Weir HK, Rosenberg HM, Vernon SW, Cronin K, Edwards BK: The annual report to the nation on the status of cancer, 1973-1997, with a special section on colorectal cancer. Cancer. 2000, 88 (10): 2398-2424. 10.1002/(SICI)1097-0142(20000515)88:10<2398::AID-CNCR26>3.0.CO;2-I.CrossRefPubMed
5.
Zurück zum Zitat Qiu Y, Langman MJ, Eggo MC: Targets of 17beta-oestradiol-induced apoptosis in colon cancer cells: a mechanism for the protective effects of hormone replacement therapy?. J Endocrinol. 2004, 181 (2): 327-337. 10.1677/joe.0.1810327.CrossRefPubMed Qiu Y, Langman MJ, Eggo MC: Targets of 17beta-oestradiol-induced apoptosis in colon cancer cells: a mechanism for the protective effects of hormone replacement therapy?. J Endocrinol. 2004, 181 (2): 327-337. 10.1677/joe.0.1810327.CrossRefPubMed
6.
Zurück zum Zitat Nelson HD, Humphrey LL, Nygren P, Teutsch SM, Allan JD: Postmenopausal hormone replacement therapy: scientific review. Jama. 2002, 288 (7): 872-881. 10.1001/jama.288.7.872.CrossRefPubMed Nelson HD, Humphrey LL, Nygren P, Teutsch SM, Allan JD: Postmenopausal hormone replacement therapy: scientific review. Jama. 2002, 288 (7): 872-881. 10.1001/jama.288.7.872.CrossRefPubMed
7.
Zurück zum Zitat Rennert G, Rennert HS, Pinchev M, Lavie O, Gruber SB: Use of hormone replacement therapy and the risk of colorectal cancer. J Clin Oncol. 2009, 27 (27): 4542-4547. 10.1200/JCO.2009.22.0764.CrossRefPubMedPubMedCentral Rennert G, Rennert HS, Pinchev M, Lavie O, Gruber SB: Use of hormone replacement therapy and the risk of colorectal cancer. J Clin Oncol. 2009, 27 (27): 4542-4547. 10.1200/JCO.2009.22.0764.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Simpson ER: Sources of estrogen and their importance. J Steroid Biochem Mol Biol. 2003, 86 (3-5): 225-230. 10.1016/S0960-0760(03)00360-1.CrossRefPubMed Simpson ER: Sources of estrogen and their importance. J Steroid Biochem Mol Biol. 2003, 86 (3-5): 225-230. 10.1016/S0960-0760(03)00360-1.CrossRefPubMed
9.
Zurück zum Zitat Mindnich R, Moller G, Adamski J: The role of 17 beta-hydroxysteroid dehydrogenases. Mol Cell Endocrinol. 2004, 218 (1-2): 7-20. 10.1016/j.mce.2003.12.006.CrossRefPubMed Mindnich R, Moller G, Adamski J: The role of 17 beta-hydroxysteroid dehydrogenases. Mol Cell Endocrinol. 2004, 218 (1-2): 7-20. 10.1016/j.mce.2003.12.006.CrossRefPubMed
10.
Zurück zum Zitat Luu The V, Labrie C, Zhao HF, Couet J, Lachance Y, Simard J, Leblanc G, Cote J, Berube D, Gagne R, et al: Characterization of cDNAs for human estradiol 17 beta-dehydrogenase and assignment of the gene to chromosome 17: evidence of two mRNA species with distinct 5'-termini in human placenta. Mol Endocrinol. 1989, 3 (8): 1301-1309. 10.1210/mend-3-8-1301.CrossRefPubMed Luu The V, Labrie C, Zhao HF, Couet J, Lachance Y, Simard J, Leblanc G, Cote J, Berube D, Gagne R, et al: Characterization of cDNAs for human estradiol 17 beta-dehydrogenase and assignment of the gene to chromosome 17: evidence of two mRNA species with distinct 5'-termini in human placenta. Mol Endocrinol. 1989, 3 (8): 1301-1309. 10.1210/mend-3-8-1301.CrossRefPubMed
11.
Zurück zum Zitat Luu-The V, Labrie C, Simard J, Lachance Y, Zhao HF, Couet J, Leblanc G, Labrie F: Structure of two in tandem human 17 beta-hydroxysteroid dehydrogenase genes. Mol Endocrinol. 1990, 4 (2): 268-275. 10.1210/mend-4-2-268.CrossRefPubMed Luu-The V, Labrie C, Simard J, Lachance Y, Zhao HF, Couet J, Leblanc G, Labrie F: Structure of two in tandem human 17 beta-hydroxysteroid dehydrogenase genes. Mol Endocrinol. 1990, 4 (2): 268-275. 10.1210/mend-4-2-268.CrossRefPubMed
12.
Zurück zum Zitat Miettinen MM, Mustonen MV, Poutanen MH, Isomaa VV, Vihko RK: Human 17 beta-hydroxysteroid dehydrogenase type 1 and type 2 isoenzymes have opposite activities in cultured cells and characteristic cell- and tissue-specific expression. Biochem J. 1996, 314 (Pt 3): 839-845.CrossRefPubMedPubMedCentral Miettinen MM, Mustonen MV, Poutanen MH, Isomaa VV, Vihko RK: Human 17 beta-hydroxysteroid dehydrogenase type 1 and type 2 isoenzymes have opposite activities in cultured cells and characteristic cell- and tissue-specific expression. Biochem J. 1996, 314 (Pt 3): 839-845.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Poutanen M, Moncharmont B, Vihko R: 17 beta-hydroxysteroid dehydrogenase gene expression in human breast cancer cells: regulation of expression by a progestin. Cancer Res. 1992, 52 (2): 290-294.PubMed Poutanen M, Moncharmont B, Vihko R: 17 beta-hydroxysteroid dehydrogenase gene expression in human breast cancer cells: regulation of expression by a progestin. Cancer Res. 1992, 52 (2): 290-294.PubMed
14.
Zurück zum Zitat Kondo Y: Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J. 2009, 50 (4): 455-463. 10.3349/ymj.2009.50.4.455.CrossRefPubMedPubMedCentral Kondo Y: Epigenetic cross-talk between DNA methylation and histone modifications in human cancers. Yonsei Med J. 2009, 50 (4): 455-463. 10.3349/ymj.2009.50.4.455.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Luczak MW, Jagodzinski PP: The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006, 44 (3): 143-154.PubMed Luczak MW, Jagodzinski PP: The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006, 44 (3): 143-154.PubMed
16.
Zurück zum Zitat Chomczynski P, Sacchi N: Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987, 162 (1): 156-159.CrossRefPubMed Chomczynski P, Sacchi N: Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Biochem. 1987, 162 (1): 156-159.CrossRefPubMed
17.
Zurück zum Zitat Radonic A, Thulke S, Mackay IM, Landt O, Siegert W, Nitsche A: Guideline to reference gene selection for quantitative real-time PCR. Biochem Biophys Res Commun. 2004, 313 (4): 856-862. 10.1016/j.bbrc.2003.11.177.CrossRefPubMed Radonic A, Thulke S, Mackay IM, Landt O, Siegert W, Nitsche A: Guideline to reference gene selection for quantitative real-time PCR. Biochem Biophys Res Commun. 2004, 313 (4): 856-862. 10.1016/j.bbrc.2003.11.177.CrossRefPubMed
18.
Zurück zum Zitat Szabo A, Perou CM, Karaca M, Perreard L, Palais R, Quackenbush JF, Bernard PS: Statistical modeling for selecting housekeeper genes. Genome Biol. 2004, 5 (8): R59-10.1186/gb-2004-5-8-r59.CrossRefPubMedPubMedCentral Szabo A, Perou CM, Karaca M, Perreard L, Palais R, Quackenbush JF, Bernard PS: Statistical modeling for selecting housekeeper genes. Genome Biol. 2004, 5 (8): R59-10.1186/gb-2004-5-8-r59.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T: BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinforma (Oxford, England). 2005, 21 (21): 4067-4068. 10.1093/bioinformatics/bti652.CrossRef Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T: BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinforma (Oxford, England). 2005, 21 (21): 4067-4068. 10.1093/bioinformatics/bti652.CrossRef
23.
Zurück zum Zitat Rohde C, Zhang Y, Jurkowski TP, Stamerjohanns H, Reinhardt R, Jeltsch A: Bisulfite sequencing Data Presentation and Compilation (BDPC) web server--a useful tool for DNA methylation analysis. Nucleic Acids Res. 2008, 36 (5): e34-10.1093/nar/gkn083.CrossRefPubMedPubMedCentral Rohde C, Zhang Y, Jurkowski TP, Stamerjohanns H, Reinhardt R, Jeltsch A: Bisulfite sequencing Data Presentation and Compilation (BDPC) web server--a useful tool for DNA methylation analysis. Nucleic Acids Res. 2008, 36 (5): e34-10.1093/nar/gkn083.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Wojdacz TK, Dobrovic A: Methylation-sensitive high resolution melting (MS-HRM): a new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res. 2007, 35 (6): e41-10.1093/nar/gkm013.CrossRefPubMedPubMedCentral Wojdacz TK, Dobrovic A: Methylation-sensitive high resolution melting (MS-HRM): a new approach for sensitive and high-throughput assessment of methylation. Nucleic Acids Res. 2007, 35 (6): e41-10.1093/nar/gkm013.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kim YI: Nutritional epigenetics: impact of folate deficiency on DNA methylation and colon cancer susceptibility. J Nutr. 2005, 135 (11): 2703-2709.PubMed Kim YI: Nutritional epigenetics: impact of folate deficiency on DNA methylation and colon cancer susceptibility. J Nutr. 2005, 135 (11): 2703-2709.PubMed
27.
Zurück zum Zitat Hermann S, Rohrmann S, Linseisen J: Lifestyle factors, obesity and the risk of colorectal adenomas in EPIC-Heidelberg. Cancer Causes Control. 2009, 20 (8): 1397-1408. 10.1007/s10552-009-9366-3.CrossRefPubMed Hermann S, Rohrmann S, Linseisen J: Lifestyle factors, obesity and the risk of colorectal adenomas in EPIC-Heidelberg. Cancer Causes Control. 2009, 20 (8): 1397-1408. 10.1007/s10552-009-9366-3.CrossRefPubMed
28.
Zurück zum Zitat van Duijnhoven FJ, Bueno-De-Mesquita HB, Ferrari P, Jenab M, Boshuizen HC, Ros MM, Casagrande C, Tjonneland A, Olsen A, Overvad K, et al: Fruit, vegetables, and colorectal cancer risk: the European Prospective Investigation into Cancer and Nutrition. American J Clin Nutr. 2009, 89 (5): 1441-1452. 10.3945/ajcn.2008.27120.CrossRef van Duijnhoven FJ, Bueno-De-Mesquita HB, Ferrari P, Jenab M, Boshuizen HC, Ros MM, Casagrande C, Tjonneland A, Olsen A, Overvad K, et al: Fruit, vegetables, and colorectal cancer risk: the European Prospective Investigation into Cancer and Nutrition. American J Clin Nutr. 2009, 89 (5): 1441-1452. 10.3945/ajcn.2008.27120.CrossRef
29.
Zurück zum Zitat Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 1990, 61 (5): 759-767. 10.1016/0092-8674(90)90186-I.CrossRefPubMed Fearon ER, Vogelstein B: A genetic model for colorectal tumorigenesis. Cell. 1990, 61 (5): 759-767. 10.1016/0092-8674(90)90186-I.CrossRefPubMed
30.
Zurück zum Zitat Kim MS, Lee J, Sidransky D: DNA methylation markers in colorectal cancer. Cancer Metastasis Rev. 2010, 29 (1): 181-206. 10.1007/s10555-010-9207-6.CrossRefPubMed Kim MS, Lee J, Sidransky D: DNA methylation markers in colorectal cancer. Cancer Metastasis Rev. 2010, 29 (1): 181-206. 10.1007/s10555-010-9207-6.CrossRefPubMed
31.
Zurück zum Zitat Miyakura Y, Sugano K, Konishi F, Ichikawa A, Maekawa M, Shitoh K, Igarashi S, Kotake K, Koyama Y, Nagai H: Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterol. 2001, 121 (6): 1300-1309. 10.1053/gast.2001.29616.CrossRef Miyakura Y, Sugano K, Konishi F, Ichikawa A, Maekawa M, Shitoh K, Igarashi S, Kotake K, Koyama Y, Nagai H: Extensive methylation of hMLH1 promoter region predominates in proximal colon cancer with microsatellite instability. Gastroenterol. 2001, 121 (6): 1300-1309. 10.1053/gast.2001.29616.CrossRef
32.
Zurück zum Zitat Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, Hubbell FA, Ascensao J, Rodabough RJ, Rosenberg CA, Taylor VM, Harris R, Chen C, et al: Estrogen plus progestin and colorectal cancer in postmenopausal women. New England J Med. 2004, 350 (10): 991-1004. 10.1056/NEJMoa032071.CrossRef Chlebowski RT, Wactawski-Wende J, Ritenbaugh C, Hubbell FA, Ascensao J, Rodabough RJ, Rosenberg CA, Taylor VM, Harris R, Chen C, et al: Estrogen plus progestin and colorectal cancer in postmenopausal women. New England J Med. 2004, 350 (10): 991-1004. 10.1056/NEJMoa032071.CrossRef
33.
Zurück zum Zitat Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, et al: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002, 288 (3): 321-333. 10.1001/jama.288.3.321.CrossRefPubMed Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ, Kooperberg C, Stefanick ML, Jackson RD, Beresford SA, Howard BV, Johnson KC, et al: Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal results From the Women's Health Initiative randomized controlled trial. JAMA. 2002, 288 (3): 321-333. 10.1001/jama.288.3.321.CrossRefPubMed
34.
Zurück zum Zitat Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Strom A, Gustafsson JA: Tumor repressive functions of estrogen receptor beta in SW480 colon cancer cells. Cancer Res. 2009, 69 (15): 6100-6106. 10.1158/0008-5472.CAN-09-0506.CrossRefPubMed Hartman J, Edvardsson K, Lindberg K, Zhao C, Williams C, Strom A, Gustafsson JA: Tumor repressive functions of estrogen receptor beta in SW480 colon cancer cells. Cancer Res. 2009, 69 (15): 6100-6106. 10.1158/0008-5472.CAN-09-0506.CrossRefPubMed
35.
Zurück zum Zitat Castiglione F, Taddei A, Degl'Innocenti DR, Buccoliero AM, Bechi P, Garbini F, Chiara FG, Moncini D, Cavallina G, Marascio L, et al: Expression of estrogen receptor beta in colon cancer progression. Diagn Mol Pathol. 2008, 17 (4): 231-236. 10.1097/PDM.0b013e3181656d67.CrossRefPubMed Castiglione F, Taddei A, Degl'Innocenti DR, Buccoliero AM, Bechi P, Garbini F, Chiara FG, Moncini D, Cavallina G, Marascio L, et al: Expression of estrogen receptor beta in colon cancer progression. Diagn Mol Pathol. 2008, 17 (4): 231-236. 10.1097/PDM.0b013e3181656d67.CrossRefPubMed
36.
Zurück zum Zitat Weige CC, Allred KF, Allred CD: Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res. 2009, 69 (23): 9118-9124. 10.1158/0008-5472.CAN-09-2348.CrossRefPubMed Weige CC, Allred KF, Allred CD: Estradiol alters cell growth in nonmalignant colonocytes and reduces the formation of preneoplastic lesions in the colon. Cancer Res. 2009, 69 (23): 9118-9124. 10.1158/0008-5472.CAN-09-2348.CrossRefPubMed
37.
Zurück zum Zitat Zhao C, Dahlman-Wright K, Gustafsson JA: Estrogen receptor beta: an overview and update. Nucl Recept signal. 2008, 6: e003-PubMedPubMedCentral Zhao C, Dahlman-Wright K, Gustafsson JA: Estrogen receptor beta: an overview and update. Nucl Recept signal. 2008, 6: e003-PubMedPubMedCentral
38.
Zurück zum Zitat Papaxoinis K, Triantafyllou K, Sasco AJ, Nicolopoulou-Stamati P, Ladas SD: Subsite-specific differences of estrogen receptor beta expression in the normal colonic epithelium: implications for carcinogenesis and colorectal cancer epidemiology. Eur J Gastroenterol Hepatol. 2010, 22 (5): 614-619. 10.1097/MEG.0b013e328335ef50.CrossRefPubMed Papaxoinis K, Triantafyllou K, Sasco AJ, Nicolopoulou-Stamati P, Ladas SD: Subsite-specific differences of estrogen receptor beta expression in the normal colonic epithelium: implications for carcinogenesis and colorectal cancer epidemiology. Eur J Gastroenterol Hepatol. 2010, 22 (5): 614-619. 10.1097/MEG.0b013e328335ef50.CrossRefPubMed
39.
Zurück zum Zitat Issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE, Baylin SB: Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet. 1994, 7 (4): 536-540. 10.1038/ng0894-536.CrossRefPubMed Issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE, Baylin SB: Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet. 1994, 7 (4): 536-540. 10.1038/ng0894-536.CrossRefPubMed
40.
Zurück zum Zitat McCashland TM, Brand R, Lyden E, de Garmo P: Gender differences in colorectal polyps and tumors. Am J Gastroenterol. 2001, 96 (3): 882-886. 10.1111/j.1572-0241.2001.03638.x.CrossRefPubMed McCashland TM, Brand R, Lyden E, de Garmo P: Gender differences in colorectal polyps and tumors. Am J Gastroenterol. 2001, 96 (3): 882-886. 10.1111/j.1572-0241.2001.03638.x.CrossRefPubMed
41.
Zurück zum Zitat Sarli L, Michiara M, Sgargi P, Iusco D, De Lisi V, Leonardi F, Bella MA, Sgobba G, Roncoroni L: The changing distribution and survival of colorectal carcinoma: an epidemiological study in an area of northern Italy. Eur J Gastroenterol Hepatol. 2005, 17 (5): 567-572. 10.1097/00042737-200505000-00014.CrossRefPubMed Sarli L, Michiara M, Sgargi P, Iusco D, De Lisi V, Leonardi F, Bella MA, Sgobba G, Roncoroni L: The changing distribution and survival of colorectal carcinoma: an epidemiological study in an area of northern Italy. Eur J Gastroenterol Hepatol. 2005, 17 (5): 567-572. 10.1097/00042737-200505000-00014.CrossRefPubMed
42.
Zurück zum Zitat Cho NL, Javid SH, Carothers AM, Redston M, Bertagnolli MM: Estrogen receptors alpha and beta are inhibitory modifiers of Apc-dependent tumorigenesis in the proximal colon of Min/+ mice. Cancer Res. 2007, 67 (5): 2366-2372. 10.1158/0008-5472.CAN-06-3026.CrossRefPubMed Cho NL, Javid SH, Carothers AM, Redston M, Bertagnolli MM: Estrogen receptors alpha and beta are inhibitory modifiers of Apc-dependent tumorigenesis in the proximal colon of Min/+ mice. Cancer Res. 2007, 67 (5): 2366-2372. 10.1158/0008-5472.CAN-06-3026.CrossRefPubMed
43.
Zurück zum Zitat Fiorelli G, Picariello L, Martineti V, Tognarini I, Tonelli F, Brandi ML: Estrogen metabolism in human colorectal cancer cells. J Steroid Biochem Mol Biol. 2002, 81 (3): 281-289. 10.1016/S0960-0760(02)00075-4.CrossRefPubMed Fiorelli G, Picariello L, Martineti V, Tognarini I, Tonelli F, Brandi ML: Estrogen metabolism in human colorectal cancer cells. J Steroid Biochem Mol Biol. 2002, 81 (3): 281-289. 10.1016/S0960-0760(02)00075-4.CrossRefPubMed
44.
Zurück zum Zitat Hanahan D, Weinberg RA: The hallmarks of cancer. Cell. 2000, 100 (1): 57-70. 10.1016/S0092-8674(00)81683-9.CrossRefPubMed Hanahan D, Weinberg RA: The hallmarks of cancer. Cell. 2000, 100 (1): 57-70. 10.1016/S0092-8674(00)81683-9.CrossRefPubMed
45.
Zurück zum Zitat Chan AO, Broaddus RR, Houlihan PS, Issa JP, Hamilton SR, Rashid A: CpG island methylation in aberrant crypt foci of the colorectum. Am J Pathol. 2002, 160 (5): 1823-1830. 10.1016/S0002-9440(10)61128-5.CrossRefPubMedPubMedCentral Chan AO, Broaddus RR, Houlihan PS, Issa JP, Hamilton SR, Rashid A: CpG island methylation in aberrant crypt foci of the colorectum. Am J Pathol. 2002, 160 (5): 1823-1830. 10.1016/S0002-9440(10)61128-5.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Suehiro Y, Hinoda Y: Genetic and epigenetic changes in aberrant crypt foci and serrated polyps. Cancer Sci. 2008, 99 (6): 1071-1076. 10.1111/j.1349-7006.2008.00784.x.CrossRefPubMed Suehiro Y, Hinoda Y: Genetic and epigenetic changes in aberrant crypt foci and serrated polyps. Cancer Sci. 2008, 99 (6): 1071-1076. 10.1111/j.1349-7006.2008.00784.x.CrossRefPubMed
47.
Zurück zum Zitat Qu Z, Yan P, Fu J, Jiang J, Grusby MJ, Smithgall TE, Xiao G: DNA methylation-dependent repression of PDZ-LIM domain-containing protein 2 in colon cancer and its role as a potential therapeutic target. Cancer Res. 2010, 70 (5): 1766-1772. 10.1158/0008-5472.CAN-09-3263.CrossRefPubMedPubMedCentral Qu Z, Yan P, Fu J, Jiang J, Grusby MJ, Smithgall TE, Xiao G: DNA methylation-dependent repression of PDZ-LIM domain-containing protein 2 in colon cancer and its role as a potential therapeutic target. Cancer Res. 2010, 70 (5): 1766-1772. 10.1158/0008-5472.CAN-09-3263.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Belanger AS, Tojcic J, Harvey M, Guillemette C: Regulation of UGT1A1 and HNF1 transcription factor gene expression by DNA methylation in colon cancer cells. BMC mol biol. 2010, 11: 9-10.1186/1471-2199-11-9.CrossRefPubMedPubMedCentral Belanger AS, Tojcic J, Harvey M, Guillemette C: Regulation of UGT1A1 and HNF1 transcription factor gene expression by DNA methylation in colon cancer cells. BMC mol biol. 2010, 11: 9-10.1186/1471-2199-11-9.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Ghoshal K, Motiwala T, Claus R, Yan P, Kutay H, Datta J, Majumder S, Bai S, Majumder A, Huang T, et al: HOXB13, a target of DNMT3B, is methylated at an upstream CpG island, and functions as a tumor suppressor in primary colorectal tumors. PloS one. 2010, 5 (4): e10338-10.1371/journal.pone.0010338.CrossRefPubMedPubMedCentral Ghoshal K, Motiwala T, Claus R, Yan P, Kutay H, Datta J, Majumder S, Bai S, Majumder A, Huang T, et al: HOXB13, a target of DNMT3B, is methylated at an upstream CpG island, and functions as a tumor suppressor in primary colorectal tumors. PloS one. 2010, 5 (4): e10338-10.1371/journal.pone.0010338.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Wendt MK, Johanesen PA, Kang-Decker N, Binion DG, Shah V, Dwinell MB: Silencing of epithelial CXCL12 expression by DNA hypermethylation promotes colonic carcinoma metastasis. Oncogene. 2006, 25 (36): 4986-4997. 10.1038/sj.onc.1209505.CrossRefPubMedPubMedCentral Wendt MK, Johanesen PA, Kang-Decker N, Binion DG, Shah V, Dwinell MB: Silencing of epithelial CXCL12 expression by DNA hypermethylation promotes colonic carcinoma metastasis. Oncogene. 2006, 25 (36): 4986-4997. 10.1038/sj.onc.1209505.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Jass JR: Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathol. 2007, 50 (1): 113-130. 10.1111/j.1365-2559.2006.02549.x.CrossRef Jass JR: Classification of colorectal cancer based on correlation of clinical, morphological and molecular features. Histopathol. 2007, 50 (1): 113-130. 10.1111/j.1365-2559.2006.02549.x.CrossRef
52.
Zurück zum Zitat Glebov OK, Rodriguez LM, Nakahara K, Jenkins J, Cliatt J, Humbyrd CJ, DeNobile J, Soballe P, Simon R, Wright G, et al: Distinguishing right from left colon by the pattern of gene expression. Cancer Epidemiol Biomarkers Prev. 2003, 12 (8): 755-762.PubMed Glebov OK, Rodriguez LM, Nakahara K, Jenkins J, Cliatt J, Humbyrd CJ, DeNobile J, Soballe P, Simon R, Wright G, et al: Distinguishing right from left colon by the pattern of gene expression. Cancer Epidemiol Biomarkers Prev. 2003, 12 (8): 755-762.PubMed
53.
Zurück zum Zitat Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP: CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999, 96 (15): 8681-8686. 10.1073/pnas.96.15.8681.CrossRefPubMedPubMedCentral Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP: CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci USA. 1999, 96 (15): 8681-8686. 10.1073/pnas.96.15.8681.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Leibovitz A, Stinson JC, McCombs WB, McCoy CE, Mazur KC, Mabry ND: Classification of human colorectal adenocarcinoma cell lines. Cancer Res. 1976, 36 (12): 4562-4569.PubMed Leibovitz A, Stinson JC, McCombs WB, McCoy CE, Mazur KC, Mabry ND: Classification of human colorectal adenocarcinoma cell lines. Cancer Res. 1976, 36 (12): 4562-4569.PubMed
55.
Zurück zum Zitat von Kleist S, Chany E, Burtin P, King M, Fogh J: Immunohistology of the antigenic pattern of a continuous cell line from a human colon tumor. J Natl Cancer Inst. 1975, 55 (3): 555-560.PubMed von Kleist S, Chany E, Burtin P, King M, Fogh J: Immunohistology of the antigenic pattern of a continuous cell line from a human colon tumor. J Natl Cancer Inst. 1975, 55 (3): 555-560.PubMed
56.
Zurück zum Zitat Site UGB: UCSC Genome Bioinformatics Site. Site UGB: UCSC Genome Bioinformatics Site.
57.
Zurück zum Zitat Lorincz MC, Dickerson DR, Schmitt M, Groudine M: Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol. 2004, 11 (11): 1068-1075. 10.1038/nsmb840.CrossRefPubMed Lorincz MC, Dickerson DR, Schmitt M, Groudine M: Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells. Nat Struct Mol Biol. 2004, 11 (11): 1068-1075. 10.1038/nsmb840.CrossRefPubMed
58.
Zurück zum Zitat Tian XQ, Zhang Y, Sun D, Zhao S, Xiong H, Fang J: Epigenetic silencing of LRRC3B in colorectal cancer. Scand J gastroenterol. 2009, 44 (1): 79-84. 10.1080/00365520802400834.CrossRefPubMed Tian XQ, Zhang Y, Sun D, Zhao S, Xiong H, Fang J: Epigenetic silencing of LRRC3B in colorectal cancer. Scand J gastroenterol. 2009, 44 (1): 79-84. 10.1080/00365520802400834.CrossRefPubMed
59.
Zurück zum Zitat Liu M, Peng Y, Wang X, Guo Q, Shen S, Li G: NGX6 gene mediated by promoter methylation as a potential molecular marker in colorectal cancer. BMC Cancer. 2010, 10: 160-10.1186/1471-2407-10-160.CrossRefPubMedPubMedCentral Liu M, Peng Y, Wang X, Guo Q, Shen S, Li G: NGX6 gene mediated by promoter methylation as a potential molecular marker in colorectal cancer. BMC Cancer. 2010, 10: 160-10.1186/1471-2407-10-160.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Lin J, Lai M, Huang Q, Ruan W, Ma Y, Cui J: Reactivation of IGFBP7 by DNA demethylation inhibits human colon cancer cell growth in vitro. Cancer biol ther. 2008, 7 (12): 1896-1900. 10.4161/cbt.7.12.6937.CrossRefPubMed Lin J, Lai M, Huang Q, Ruan W, Ma Y, Cui J: Reactivation of IGFBP7 by DNA demethylation inhibits human colon cancer cell growth in vitro. Cancer biol ther. 2008, 7 (12): 1896-1900. 10.4161/cbt.7.12.6937.CrossRefPubMed
Metadaten
Titel
Decreased expression of 17β-hydroxysteroid dehydrogenase type 1 is associated with DNA hypermethylation in colorectal cancer located in the proximal colon
verfasst von
Agnieszka Anna Rawłuszko
Karolina Horbacka
Piotr Krokowicz
Paweł Piotr Jagodziński
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2011
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-11-522

Weitere Artikel der Ausgabe 1/2011

BMC Cancer 1/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.