Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 5/2020

Open Access 18.03.2020 | Original Article – Cancer Research

Downregulation of SFRP1 is a protumorigenic event in hepatoblastoma and correlates with beta-catenin mutations

verfasst von: Ivonne Regel, Melanie Eichenmüller, Ujjwal Mukund Mahajan, Beate Hagl, Simone Benitz, Beate Häberle, Christian Vokuhl, Dietrich von Schweinitz, Roland Kappler

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 5/2020

Abstract

Background

Hepatoblastoma (HB) and pediatric hepatocellular carcinoma (HCC) are the most common malignant liver tumors in childhood. Both tumor types exhibit genetic and epigenetic alterations in the WNT/β-catenin signaling pathway, which is a key regulator of liver progenitor cells in embryonic development. The tumors demonstrate a high rate of β-catenin mutations and gene expression changes of several WNT antagonists. However, the role of the WNT inhibitory factor secreted frizzled-related protein 1 (SFRP1) has not been addressed in pediatric liver cancer so far.

Results

In our study, we investigated the gene expression level, DNA methylation status and functional relevance of SFRP1 in HB cell lines and in pediatric liver tumor patient samples. SFRP1 was downregulated due to DNA promoter methylation in all tested HB cell lines. Overexpression of SFRP1 in HB cell lines diminished tumor cell proliferation, colony formation and migration potential. In addition, the SFRP1-expressing HB cell lines showed reduced WNT/β-catenin signaling pathway activity and decreased expression of WNT target genes. To evaluate the utility of SFRP1 as a biomarker in pediatric liver cancer, we determined the gene expression level and DNA methylation status of SFRP1 in 45 pediatric liver tumor patient samples. The correlation analysis of different clinical parameters and tumor characteristics revealed a significant correlation of reduced SFRP1 expression with the presence of mutant β-catenin. The methylation status of SFRP1 was furthermore associated to a pediatric liver tumor type with HCC-like characteristics, TERT mutations and an older age at diagnosis.

Conclusion

Altogether, our data demonstrate that the epigenetic suppression of the WNT/β-catenin antagonist SFRP1 has an important impact on the malignant behavior of HB cells. Although SFRP1 methylation is a common event in HCC-like pediatric liver tumors, its potential as a prognostic or diagnostic biomarker needs to be further investigated.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s00432-020-03182-1) contains supplementary material, which is available to authorized users.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
HB
Hepatoblastoma
HCC
Hepatocellular carcinoma
NL
Normal pediatric liver tissue
MSP
Methylation-specific-PCR
5-aza
5-aza-2′-deoxycytidine
pSFRP1
PcDNA3.1-SFRP1 plasmid
TLCT
Transitional liver cell tumors
NSET
Nested stromal-epithelial liver tumors
PRETEXT
PRE-Treatment EXTent of tumor
EGCG
Epigallocatechin-3-gallate
m
Male
f
Female
DOD
Died of disease
NED
No evidence of disease
E
Embryonal
F
Fetal
pure OS
Pure osteoid
C1 and C2
16-gene signature cluster 1 and 2 (Cairo et al. 2008)
M
Methylated
U
Unmethylated
nd
No data
na
Not applicable

Background

Hepatoblastoma (HB) is the most common malignant liver tumor in children under the age of 4 years and its incidence has increased over the last decades (Kremer et al. 2014). Hepatocellular carcinoma (HCC) has also been described in the pediatric population, but the tumor occurs more rarely than HB and predominantly manifest in older children or young adults (Ng and Mogul 2018). Although the survival rates of pediatric HB and HCC have improved up to 80% over the last decades, due to advanced surgical techniques and chemotherapeutic treatments, there are still HB patient subgroups with a dismal prognosis (Tulla et al. 2015; Khanna and Verma 2018; Czauderna and Garnier 2018). Particularly, children presenting with non-resectable tumors, chemotherapy resistance, or metastasis show reduced survival rates (Czauderna et al. 2016). Consequently, a more profound understanding of the biology of HB will help to uncover mechanisms promoting cancer development and progression.
HBs often exhibit an epithelial or mixed morphological subtype and show two distinct transcriptomic profiles, which are classified as 16-gene signature cluster C1 or C2 (Perugorria et al. 2019). Although C1 and C2 HBs differ in their tumor characteristics, they exhibit common genetic aberrations affecting the WNT/β-catenin/AXIN signaling (Cairo et al. 2008). The canonical WNT/β-catenin signaling pathway regulates the proliferation, maturation and survival of liver progenitor cells in embryogenesis. Notably, the pathway is often constitutively activated in HB and HCC development (Russell and Monga 2018). Stabilizing mutations in the β-catenin (CTNNB1) gene occur with a frequency of up to 37% in HCC and 60–70% in HB and as a consequence, β-catenin accumulates in the nucleus and regulates the expression of target genes (Zucman-Rossi et al. 2015; Bell et al. 2017). Further genetic alterations involve loss-of-function mutations in the APC regulator of WNT signaling pathway (APC), AXIN1 and AXIN2 genes, which prevents a proteasomal degradation of β-catenin (Perugorria et al. 2019). Moreover, it was previously shown that the dickkopf WNT signaling pathway inhibitor 1 (DKK1) is upregulated in HB patient samples, which might represent negative feedback mechanisms (Wirths et al. 2003). On the contrary, among the known WNT antagonists, the secreted frizzled-related protein 1 (SFRP1) is often downregulated in various cancer entities, which indicates that SFRP1 has tumor-suppressive functions (Vincent and Postovit 2017). Under physiological conditions, SFRP1 inactivates the canonical and non-canonical WNT/β-catenin pathway by directly binding to WNT proteins or the frizzled receptor (Kawano and Kypta 2003). Thus, a downregulation of SFRP1 in HCC cells resulted in stimulated WNT signaling activity and increased tumor cell growth (Shih et al. 2007). Importantly, during breast and prostate cancer development SFRP1 expression is lost due to epigenetic silencing, induced by an enrichment of DNA methylation in the promoter region (Lodygin et al. 2005; Lo et al. 2006). Notably, promoter methylation of SFRP1 was also identified as a common event in adult HCC (Huang et al. 2007; Shih et al. 2006). Since SFRP1 suppression contributes to elevated WNT/β-catenin signaling, which is a known characteristic of HB and HCC, we were highly interested in the functional role of SFRP1 in pediatric liver cancers. Thus, we investigated the SFRP1 DNA methylation status and gene expression levels in HB cell lines and primary pediatric liver tumor samples. Overexpression of SFRP1 in HB cell lines resulted in an inhibition of tumor cell growth, colony formation and migration and a decrease in WNT/β-catenin signaling activity. Moreover, SFRP1 promoter methylation and transcriptional silencing was identified in a subset of primary pediatric liver tumors. Our findings indicate that the epigenetic suppression of SFRP1 represents an alternative mechanism for enhancing WNT/β-catenin signaling in the development of pediatric liver cancer, particularly in children diagnosed at older ages.

Methods

Patients

Liver tumor specimens of 45 patients and matching normal liver tissue from seven patients (N110, N146, N198, N175, N227, N253, N612) were obtained from pediatric patients undergoing surgical resection in the Department of Pediatric Surgery, University Hospital, LMU Munich, Germany. Each patient gave written informed consent and the study protocol was approved by the Committee of Ethics, LMU Munich. Clinicopathological parameters and experimental data of all patient samples are listed in the Supplementary Table 1. Experimental data of SFRP1 expression were categorized into low (< 1) and high (> 1) and correlated to different clinicopathological parameters. A similar correlation analysis was performed for the SFRP1 methylation status (M methylated, U unmethylated).

Cell culture and DNA methylation inhibitor treatment

The hepatoblastoma cell lines HuH-6 (RRID:CVCL_4381), HepT1 (RRID:CVCL_G003), Hep-T3 (RRID:CVCL_G004), and HepG2 (RRID:CVCL_0027) were cultured in RPMI 1640 growth media (Gibco, Thermo Fischer Scientific, Germany), supplemented with 10% fetal calf serum, 100 U/mL penicillin, and 100 μg/mL streptomycin, at 37 °C in a humidified chamber with a saturated atmosphere containing 5% CO2. Cells were passaged at a confluency of 80–90% with 0.05% trypsin (v/v) and 0.2% EDTA (w/v) (Sigma-Aldrich, Germany) in Dulbecco’s phosphate-buffered saline (PBS). For gene expression and methylation analysis Huh-6 and Hep-T3 cells were treated with 0.5 µM 5-aza-2′-deoxycytidine (5-aza; Sigma-Aldrich, Germany), HepT1 and HepG2 cells with 1.25 µM 5-aza or solvent for 3 and 5 days.

Cell transfection

HuH-6, HepT1 and HepG2 cells (5 × 105 cells/six-well plate) were transfected with 1 μg DNA of the pcDNA3.1/V5-HisA control vector (#V81020, Thermo Fischer Scientific, Germany) or the pcDNA3.1-SFRP1 (pSFRP1) expression vector containing full-length SFRP1 cDNA (Fukui et al. 2005) using FuGene 6 transfection reagent (Roche Diagnostics, Germany) according to the manufacturer’s protocol. For stable transfection, cells were incubated in selection media 24 h after transfection containing 200 µg/ml G418 (Sigma-Aldrich, Germany). Two weeks after G418 selection, resistant colonies were picked and cultured under standard medium conditions.

Cell viability assay

2000 stably or transiently transfected HuH-6, HepT1 and HepG2 cells were seeded in a 96-well plates in RPMI 1640 growth media and cell proliferation was measured at the indicated time points using the Cell Proliferation Kit I (Roche Diagnostics) according to the manufacturer’s protocol. The absorbance of the colorimetric reaction was quantified on the GENios reader (Tecan, Switzerland) by measuring at a wavelength of 595 nm. Cell growth was normalized to the zero hour time points.

Colony formation assay

5000 stably transfected HuH-6 and HepG2 cells were seeded in a six-well plate. Cells grew for 10 days in RPMI 1640 growth media. After methanol fixation, cells were stained with 0.05% crystal violet in 20% methanol and washed with tap water. Colonies were counted and are represented as number of colonies per well.

Cell migration assay

Stably transfected HuH-6 cells were seeded into six-well plates and grown as confluent monolayer. A wound of approximately 1 mm was inflicted to the cell monolayer using a pipette tip. The cells were washed twice with PBS to remove detached cells and incubated for additional 72 h in 1% FCS starved RPMI 1640 growth media to diminish cell proliferation. Images were taken at 0, 24, 48 and 72 h after scratching and the wound widths were measured and quantified with ImageJ (Rasband, W.S., ImageJ, US National Institutes of Health, Bethesda, USA). Cell migration was normalized to the zero hour time points.

TOP/FOP assay

To measure the activity of the canonical WNT/β-catenin pathway, we used the TOP/FOP-flash promoter assay (Millipore, Germany). 1 × 105 stably transfected HuH-6, HepT1 and HepG2 cells were seeded in a 12-well plate and co-transfected with FuGene 6 transfection reagent (Roche Diagnostics) using the following plasmids: pTOP (Firefly-Luciferase reporter plasmid containing several TCF binding sites) or pFOP (Firefly-Luciferase reporter plasmid containing mutated TCF binding sites) together with pRL-TK (Renilla-Luciferase control plasmid to normalize transfection efficiency). The luciferase activity was measured with the Dual-Glo™ Luciferase Assay System (Promega, Germany) 48 h after transfection according to the manufacturer’s protocol on the GENios microplate reader (Tecan).

Immunofluorescence staining

Staining was performed according to standard protocol using β-catenin (D10A8) antibody (#8480, cell signaling, USA). Negative control was performed with secondary antibody only (data not shown).

Immunoblot analysis

Immunoblot analysis was performed according to the manufacturer’s protocol, with the exception that blocking and first antibody (SFRP1 (D5A7) #3534, cell signaling, USA; GAPDH, H86504M, Meridian Life Science, USA) incubation was done in 5% BSA/TBS-T buffer. Secondary antibody was incubated in 5% milk/TBS-T.

Quantitative real-time PCR (qRT-PCR)

Total RNA was extracted using TriReagent (Sigma-Aldrich, Germany) according to the manufacturer’s protocol. Two micrograms of RNA was transcribed into cDNA using random hexamer primer and SuperScript™ II Reverse Transcriptase (Thermo Fischer Scientific, Germany). Quantitative RT-PCR was performed in doublets using iTaq-SYBR Green-Supermix (Bio-Rad, Germany) and the Master cycler ep gradient (Eppendorf, Germany) as previously described (Eichenmuller et al. 2009). Gene expression primer are provided in Supplementary Table 2. Expression levels were normalized to the housekeeping gene TATA-box binding protein (TBP) and the fold change was calculated according to the ΔΔCt method in relation to the expression level of normal liver tissue. An expression level < 1 represents a low and > 1 a high expression.

Methylation analyses

Genomic DNA was isolated by phenol and chloroform extraction following standard procedures. As a positive control for methylated DNA, genomic DNA of a healthy donor was artificially methylated using the CpG methyltransferase M. SssI (Thermo Fischer Scientific, Germany) according to the manufacturer's instructions. As a negative control for unmethylated DNA, HuH-6 cells were treated with 0.5 μM 5-aza-2′-deoxycytidine (5-aza; Sigma-Aldrich, Germany) for 72 h and genomic DNA was isolated. Genomic DNA was bisulfite-treated using the EpiTect® Bisulfite Kit (Qiagen, Hilden, Germany) according to the manufacturer’s protocol. The methylation status of APC, DKK1, SFRP1 and WIF1 was analyzed by methylation-specific-PCR (MSP). MSP primer are provided in Supplementary Table 2 (Aguilera et al. 2006; Esteller et al. 2000). MSP primer design and PCR conditions were previously described (Eichenmuller et al. 2009). PCR products were visualized on a 1.5% agarose gel.

Chromatin immunoprecipitation

HuH-6, HepT1 and HepG2 cells were treated with 1.5 µM 5-aza, 1.5 µM 5-aza plus 0.5 µM Vorinostat (SAHA) (SML0061, Sigma-Aldrich, Germany) or dissolvent (DMSO, 1:1000) for 3 days. Chromatin immunoprecipitation was performed as previously described (Benitz et al. 2019). Here, we used the following antibodies: anti-H3K27ac (Acetyl-Histone H3 (Lys27) (D5E4), #8173, Cell Signaling, USA), or IgG control (sc-2027, Santa Cruz Biotechnology, USA). Pull-down was done with Protein A agarose/salmon sperm DNA (Merck Millipore, Germany). After reverse crosslinking, DNA was purified with the Qiaquick® PCR Purification Kit (Qiagen) and two genomic areas around transcriptional start side of SFRP1 were amplified and quantified by qRT-PCR. Analysis of the EPCAM promoter was included as quality control to ensure specific enrichment of the activating histone modifications. Primer sequences are listed in Supplementary Table 2. Sample values were calculated according to the percent input method.

Statistical analysis

Data are presented as mean ± SEM. Statistical significance was determined by two-tailed, unpaired Student’s t test, one-way ANOVA with Dunnett’s multiple comparisons test or Chi-square test as indicated in the figure legends using GraphPad Prism 8 software (GraphPad Software Inc.) or R 2.1.0 (https://​cran.​r-project.​org/​src/​base/​R-2/​R-2.​1.​0.​tar.​gz) and R-studio Version 1.1.442. A two-sided significance of p < 0.05 was used throughout, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Results

Promoter methylation causes SFRP1 silencing in human HB cell lines

Although the WNT/β-catenin signaling pathway is often constitutively activated in liver cancer, due to stabilizing β-catenin mutations, previous data indicate that an additional suppression of the WNT antagonist, particularly of SFRP1, through epigenetic mechanisms is an important event in cancer formation (Kaur et al. 2012). To analyze the gene expression status of the known WNT antagonists APC, DKK1, SFRP1 and WIF1 in HB, we selected the HB cell lines HuH-6, HepT1, Hep-T3 and HepG2 and compared the expression levels to normal pediatric liver tissue (NL) (Fig. 1a). Here, APC and SFRP1 were downregulated in all four HB cell lines. In contrast, DKK1 and WIF1 showed a heterogeneous expression pattern, with an increased expression of DKK1 in HuH-6, HepT1 and Hep-T3 and of WIF1 in Hep-T3 cells (Fig. 1a). Next, we investigated whether the gene expression of APC, DKK1, SFRP1 and WIF1 is epigenetically controlled by DNA promoter methylation. For this, we performed methylation-specific-PCR (MSP) after bisulfite treatment of DNA isolated from HB cell lines and identified that all four HB cell lines demonstrated a strong DNA promoter methylation of the APC, DKK1, SFRP1 and WIF1 genes, with an exception for DKK1, which was unmethylated in HuH-6 cells (Fig. 1b). Treatment with the DNA methylation inhibitor 5-aza-2´-deoxycytidine (5-aza) for 3 and 5 days revealed a strong DNA demethylation of the analyzed promoter areas in all cell lines (Fig. 1b). However, 5-aza treatment did not affect APC expression. DKK1 and WIF1, which show originally a high expression in HepT1 and/or Hep-T3 cells, were unexpectedly downregulated after 5-aza treatment (Fig. 1c). Interestingly, only the demethylation of the SFRP1 promoter was associated with a consistent restoration of gene expression in all four HB cell lines (Fig. 1c). Immunoblot analysis of 5-aza-treated HuH6, HepT1 and HepG2 cells revealed increased SFRP1 expression after 3 days (Supplementary Fig. 1a). Moreover, we analyzed if other epigenetic mechanisms, such as histone acetylation, regulate SFRP1 gene expression. The 5-aza treatment of HuH-6, HepT1 and HepG2 cells alone or in combination with an HDAC inhibitor (SAHA) did not result in increased histone acetylation levels at two regulatory SFRP1 genome sites, as it could be shown for EPCAM, which was included as positive control (Supplementary Fig. 1b). In conclusion, our data suggest that solely SFRP1 promoter hypermethylation is associated with a transcriptional silencing in HB tumor cell lines.

Restored SFRP1 expression affects WNT signaling activity and HB tumor cell characteristics

To study the functional relevance of SFRP1 gene silencing in HB, we assessed tumor cell characteristics after SFRP1 re-expression. To restore SFRP1 gene expression, we transiently transfected HuH-6, HepT1 and HepG2 cells with the pcDNA3.1-SFRP1 plasmid (pSFRP1), containing full-length SFRP1 cDNA, or with the empty vector (pcDNA3.1) as control. Expression levels of SFRP1 were determined 48 and 72 h after transfection and showed markedly elevated SFRP1 transcript levels (Fig. 2a, Supplementary Fig. 2a). Notably, transiently pSFRP1-transfected HuH-6, HepT1 and HepG2 cells displayed a reduced growth rate compared to control-transfected cells (Fig. 2b, Supplementary Fig. 2b). To analyze long-term effects, we generated pSFRP1 or pcDNA3.1 stably transfected HuH-6, HepT1 and HepG2 cells. For instance, the HuH-6 cell clone 2 showed a substantial increase in SFRP1 gene and protein expression (Fig. 2c). In line with our preceding results, the stable SFRP1 expression in HuH-6, HepT1 and HepG2 cells resulted in significantly impaired tumor cell growth (Fig. 2d, Supplementary Fig. 2c) and a strong decrease in colony formation (Fig. 2e, Supplementary Fig. 2d) and migration capacity (Fig. 2f) compared to control cells. To investigate whether the re-expression of SFRP1 has a direct influence on the activity of the canonical WNT/β-catenin signaling pathway, we performed a TOP/FOP luciferase reporter assay, in which the binding activity of ß-catenin to the TOP reporter plasmids is measured. Strikingly, the stably pSFRP1-transfected HuH-6, HepT1 and HepG2 cells revealed a substantial reduction in the relative luciferase activity, indicating a suppression of the canonical WNT/β-catenin pathway activity (Fig. 2g, Supplementary Fig. 2e). To corroborate these findings, we additionally measured the expression of WNT target genes and observed a significant downregulation of MYC and CCND1 in the stably pSFRP1-transfected HuH-6 cells (Fig. 2h). Interestingly, the level of β-catenin or its cellular localization is not altered in the stably pSFRP1-transfected HuH-6 cells, indicating that SFRP1 re-expression abolishes the transcriptional activity of β-catenin (Fig. 2i). Altogether, our results demonstrate that a restored SFRP1 expression has tumor-suppressive effects in HB cells by reducing the activity of the canonical WNT/β-catenin pathway.

SFRP1 DNA methylation correlates with the tumor type and a late onset of the disease

To uncover the SFRP1 gene expression and DNA methylation status in primary pediatric liver tumors, we first performed a gene expression analysis on a cohort of 45 patient samples, containing 30 HBs, nine HCCs, 2 transitional liver cell tumors (TLCT) and 2 nested stromal-epithelial liver tumors (NSET) (Supplementary Table 1). Overall, we detected a low SFRP1 gene expression in 62% (28/45) of cases and the median SFRP1 expression level was reduced in tumor tissue compared to normal liver, although the difference was not significant (Fig. 3a, b). In addition, we determined the SFRP1 DNA methylation status for each case with MSP and correlated it to the SFRP1 gene expression level (Fig. 3b). Notably, 43% of the patient samples with low SFRP1 expression showed SFRP1 DNA methylation. Concomitantly, however, 35% of the patient samples with a high SFRP1 expression showed also SFRP1 DNA methylation. Hence, the overall correlation between the SFRP1 DNA methylation and transcriptional status was not significant (Fig. 3b). Representative results of the MSP reactions are illustrated in Fig. 3c. To study the impact of the SFRP1 gene expression level on clinical outcome, we performed a Chi-square correlation analysis based on a low (< 1) and high (> 1) SFRP1 expression and various clinicopathological parameters (Table 1). Here, we uncovered that the SFRP1 expression is significantly associated with the PRETEXT (PRE-Treatment EXTent of tumor) risk classification system (Towbin et al. 2018). Particularly, all patient samples (6/6) with the unfavorable PRETEXT category 4 displayed a low SFRP1 gene expression (Table 1). Moreover, the transcriptional level of SFRP1 correlated significantly with the ß-catenin (CTNNB1) mutation status. Interestingly, patient samples with mutant ß-catenin demonstrated a reduction in SFRP1 gene expression, whereas patients with wildtype ß-catenin revealed increased SFRP1 expression levels (Fig. 3d). Since 18 out of 45 patient samples exhibited SFRP1 DNA methylation, we performed also a Chi-square correlation analysis based on a methylated (M) and unmethylated (U) SFRP1 profile (Table 2). The SFRP1 methylation status was significantly associated with the gender, age at diagnosis, tumor type, differentiation, extrahepatic growth, resection margin and telomerase reverse transcriptase (TERT) mutations (Table 2). The distribution of SFRP1-methylated and -unmethylated cases in relation to the tumor type clearly showed that most of the HCC/TLCT samples displayed an enrichment of SFRP1 DNA methylation and that all tumors with a TERT mutation were SFRP1-methylated (Fig. 3e). Of further note, SFRP1 DNA methylation as a single parameter was significantly associated with an older age at diagnosis, although a low SFRP1 expression did not correlate with the age in a simple or cumulative correlation model (Fig. 3f). Overall, our findings revealed that SFRP1 DNA methylation and transcriptional silencing is a common event in pediatric liver cancer and that SFRP1 DNA methylation is a preferential characteristic of pediatric liver tumors with HCC-like features, such as hepatocellular histology, advanced age and TERT mutations.
Table 1
Correlation analysis of SFRP1 gene expression
SFRP1 expression
High (n = 17)
Low (n = 28)
Total (n = 45)
p value
Gender
0.848
 f
8 (47.1%)
14 (50.0%)
22 (48.9%)
 
 m
9 (52.9%)
14 (50.0%)
23 (51.1%)
 
Age at diagnosis in month
0.386
 nd
0
1
1
 
 Mean (SD)
67.867 (72.955)
50.780 (55.905)
57.382 (62.777)
 
 Range
1.874–199.435
0.000–184.179
0.000–199.435
 
Outcome
0.758
 DOD
3 (17.6%)
6 (21.4%)
9 (20.0%)
 
 NED
14 (82.4%)
22 (78.6%)
36 (80.0%)
 
Cause of death
0.541
 nd
0
1
1
 
 Progressive
2 (11.8%)
2 (7.4%)
4 (9.1%)
 
 Recurrence
2 (11.8%)
3 (11.1%)
5 (11.1%)
 
 Alive
13 (76.5%)
22 (81.5%)
35 (79.5%)
 
Tumor type
0.170
 HB
10 (58.8%)
20 (71.4%)
30 (66.7%)
 
 HCC/TLCT
5 (29.4%)
8 (28.6%)
13 (28.9%)
 
 NSET
2 (11.8%)
0 (0.0%)
2 (4.4%)
 
Differentiation
0.388
 nd
2
0
2
 
 Epithelial
6 (40.0%)
17 (60.7%)
23 (53.5%)
 
 Fibrolamellar
3 (20.0%)
1 (3.6%)
4 (9.3%)
 
 Well differentiated
0 (0.0%)
2 (7.1%)
2 (4.4%)
 
 Moderately differentiated
1 (6.7%)
2 (7.1%)
3 (7.0%)
 
 Mixed
5 (33.3%)
6 (21.4%)
11 (25.6%)
 
Component
0.332
 na
6 (35.3%)
5 (17.9%)
11 (24.4%)
 
 E
1 (5.9%)
0 (0.0%)
1 (2.2%)
 
 E/F
3 (17.6%)
6 (21.4%)
9 (20.0%)
 
 E > F
1 (5.9%)
1 (3.6%)
2 (4.4%)
 
 F
2 (11.8%)
4 (14.3%)
6 (13.3%)
 
 F > E
3 (17.6%)
12 (42.9%)
15 (33.3%)
 
 Pure OS
1 (5.9%)
0 (0.0%)
1 (2.2%)
 
Stage
0.296
 nd
2
1
3
 
 I
1 (6.7%)
6 (22.2%)
7 (16.7%)
 
 II
0 (0.0%)
1 (3.7%)
1 (2.4%)
 
 III
9 (60.0%)
9 (33.3%)
18 (42.9%)
 
 IV
5 (33.3%)
11 (40.7%)
16 (38.1%)
 
PRETEXT
0.025
 nd
3
1
4
 
 1
2 (13.3%)
0 (0.0%)
2 (4.8%)
 
 2
3 (20.0%)
11 (40.7%)
14 (33.3%)
 
 3
9 (60.0%)
10 (37.0%)
19 (45.2%)
 
 4
0 (0.0%)
6 (22.2%)
6 (14.3%)
 
Extrahepatic
0.820
 nd
0
2
2
 
 No
16 (94.1%)
24 (92.3%)
40 (93.0%)
 
 Yes
1 (5.9%)
2 (7.7%)
3 (7.0%)
 
Multifocal
0.666
 nd
2
1
3
 
 No
12 (80.0%)
20 (74.1%)
32 (76.2%)
 
 Yes
3 (20.0%)
7 (25.9%)
10 (23.8%)
 
Metastasis
0.447
 nd
0
1
1
 
 No
12 (70.6%)
16 (59.3%)
28 (63.6%)
 
 Yes
5 (29.4%)
11 (40.7%)
16 (36.4%)
 
Chemotherapy
0.282
 No
2 (11.8%)
7 (25.0%)
9 (20.0%)
 
 Yes
15 (88.2%)
21 (75.0%)
36 (80.0%)
 
Resection margin
0.911
 Nd
2
1
3
 
 R0
13 (81.2%)
23 (82.1%)
36 (81.8%)
 
 R1
2 (12.5%)
4 (14.3%)
6 (13.6%)
 
16-gene signature
0.172
 na
2 (11.8%)
0 (0.0%)
2 (4.4%)
 
 C1
9 (52.9%)
18 (64.3%)
27 (60.0%)
 
 C2
6 (35.3%)
10 (35.7%)
16 (35.6%)
 
CTNNB1
0.017
 Mutant
11 (64.7%)
26 (92.9%)
37 (82.2%)
 
 Wildtype
6 (35.3%)
2 (7.1%)
8 (17.8%)
 
NFE2L2
0.137
 nd
3
0
3
 
 Mutant
0 (0.0%)
4 (14.3%)
4 (9.5%)
 
 Wildtype
14 (100.0%)
24 (85.7%)
38 (90.5%)
 
TERT
0.620
 nd
1
0
1
 
 Mutant
1 (6.2%)
3 (10.7%)
4 (9.1%)
 
 Wildtype
15 (93.8%)
25 (89.3%)
40 (90.9%)
 
SFRP1 methylation
0.616
 M
6 (35.3%)
12 (42.9%)
18 (40.0%)
 
 U
11 (64.7%)
16 (57.1%)
27 (60.0%)
 
Chi-square correlation analysis of normalized low (< 1) and high (> 1) SFRP1 expression categories to different clinicopathological and experimental parameters
m male, f female, DOD died of disease, NED no evidence of disease, HB hepatoblastoma, HCC hepatocellular carcinoma, TLCT transitional liver cell tumor, NSET nested stromal-epithelial liver tumor, E embryonal, F fetal, pure OS pure osteoid, C1 and C2 16-gene signature cluster C1 and C2 (Cairo et al. 2008), M methylated, U unmethylated, nd no data, na not applicable
Table 2
Correlation analysis of SFRP1 promoter methylation
SFRP1 methylation
M (n = 18)
U (n = 27)
Total (n = 45)
p value
Gender
   
0.021
 f
5 (27.8%)
17 (63.0%)
22 (48.9%)
 
 m
13 (72.2%)
10 (37.0%)
23 (51.1%)
 
Age at diagnosis in month
  
 < 0.001
 nd
1
0
1
 
 Mean (SD)
98.48 (73.09)
31.50 (37.82)
57.38 (62.78)
 
 Range
7.37–199.44
0.00–156.33
0.00–199.44
 
Outcome
   
0.287
 DOD
5 (27.8%)
4 (14.8%)
9 (20.0%)
 
 NED
13 (72.2%)
23 (85.2%)
36 (80.0%)
 
Cause of death
0.512
 nd
1
0
1
 
 Progressive
2 (11.8%)
2 (7.4%)
4 (9.1%)
 
 Recurrence
3 (16.6%)
2 (7.4%)
5 (11.1%)
 
 Alive
12 (70.6%)
23 (85.2%)
35 (79.5%)
 
Tumor type
0.004
 HB
7 (38.9%)
23 (85.2%)
30 (66.7%)
 
 HCC/TLCT
10 (55.6%)
3 (11.1%)
13 (28.9%)
 
 NSET
1 (5.6%)
1 (3.7%)
2 (4.4%)
 
Differentiation
0.042
 nd
1
1
2
 
 Epithelial
8 (47.1%)
15 (57.7%)
23 (53.5%)
 
 Fibrolamellar
4 (23.5%)
0 (0.0%)
4 (9.3%)
 
 Well differentiated
1 (5.9%)
1 (3.8%)
2 (4.4%)
 
 Moderately differentiated
2 (11.8%)
1 (3.8%)
3 (7.0%)
 
 Mixed
2 (11.8%)
9 (34.6%)
11 (25.6%)
 
Component
0.260
 na
8 (44.4%)
3 (11.1%)
11 (24.4%)
 
 E
0 (0.0%)
1 (3.7%)
1 (2.2%)
 
 E/F
3 (16.7%)
6 (22.2%)
9 (20.0%)
 
 E > F
1 (5.6%)
1 (3.7%)
2 (4.4%)
 
 F
2 (11.1%)
4 (14.8%)
6 (13.3%)
 
 F > E
4 (22.2%)
11 (40.7%)
15 (33.3%)
 
 pure OS
0 (0.0%)
1 (3.7%)
1 (2.2%)
 
Stage
0.078
 nd
1
2
3
 
 I
5 (29.4%)
2 (8.0%)
7 (16.7%)
 
 II
1 (5.9%)
0 (0.0%)
1 (2.4%)
 
 III
4 (23.5%)
14 (56.0%)
18 (42.9%)
 
 IV
7 (41.2%)
9 (36.0%)
16 (38.1%)
 
PRETEXT
0.552
 nd
4
0
4
 
 1
1 (6.7%)
1 (3.7%)
2 (4.8%)
 
 2
6 (40.0%)
8 (29.6%)
14 (33.3%)
 
 3
5 (33.3%)
14 (51.9%)
19 (45.2%)
 
 4
2 (13.3%)
4 (14.8%)
6 (14.3%)
 
Extrahepatic
0.020
 nd
2
0
2
 
 No
13 (81.2%)
27 (100.0%)
40 (93.0%)
 
 Yes
3 (18.8%)
0 (0.0%)
3 (7.0%)
 
Multifocal
0.280
 nd
3
0
3
 
 No
10 (66.7%)
22 (81.5%)
32 (76.2%)
 
 Yes
5 (33.3%)
5 (18.5%)
10 (23.8%)
 
Metastasis
0.598
 nd
1
0
1
 
 No
10 (58.8%)
18 (66.7%)
28 (63.6%)
 
 Yes
7 (41.2%)
9 (33.3%)
16 (36.4%)
 
Chemotherapy
0.761
 No
4 (22.2%)
5 (18.5%)
9 (20.0%)
 
 Yes
14 (77.8%)
22 (81.5%)
36 (80.0%)
 
Resection margin
0.046
 nd
3
0
3
 
 R0
11 (64.7%)
25 (92.6%)
36 (81.8%)
 
 R1
4 (23.5%)
2 (7.4%)
6 (13.6%)
 
16-gene signature
0.871
 na
1 (5.6%)
1 (3.7%)
2 (4.4%)
 
 C1
10 (55.6%)
17 (63.0%)
27 (60.0%)
 
 C2
7 (38.9%)
9 (33.3%)
16 (35.6%)
 
CTNNB1
0.340
 Mutant
16 (88.9%)
21 (77.8%)
37 (82.2%)
 
 Wildtype
2 (11.1%)
6 (22.2%)
8 (17.8%)
 
NFE2L2
0.571
 nd
2
1
3
 
 Mutant
1 (6.2%)
3 (11.5%)
4 (9.5%)
 
 Wildtype
15 (93.8%)
23 (88.5%)
38 (90.5%)
 
TERT
0.008
 nd
1
0
1
 
 Mutant
4 (23.5%)
0 (0.0%)
4 (9.1%)
 
 Wildtype
13 (76.5%)
27 (100.0%)
40 (90.9%)
 
SFRP1 expression
0.908
 Mean (SD)
1.327 (1.956)
1.265 (1.575)
1.290 (1.716)
 
 Range
0.015–6.287
0.029–6.450
0.015–6.450
 
SFRP1 exp. category
0.616
 High
6 (33.3%)
11 (40.7%)
17 (37.8%)
 
 Low
12 (66.7%)
16 (59.3%)
28 (62.2%)
 
Chi-square correlation analysis of methylated (M) and unmethylated (U) SFRP1 categories to different clinicopathological and experimental parameters
m male, f female, DOD died of disease, NED no evidence of disease, HB hepatoblastoma, HCC hepatocellular carcinoma, TLCT transitional liver cell tumor, NSET nested stromal-epithelial liver tumor, E embryonal, F fetal, pure OS pure osteoid, C1 and C2 16-gene signature cluster C1 and C2 (Cairo et al. 2008), nd no data, na not applicable

Discussion

A constitutive activation of the WNT/β-catenin signaling pathway is a common event in pediatric liver tumor development. During embryogenesis, the pathway controls liver development and hepatoblast proliferation, indicating that a signaling malfunction contributes to liver cell transformation and tumor development (Perugorria et al. 2019). Indeed, HBs and HCCs exhibit an increased mutational burden in WNT/β-catenin pathway components that affect CTNNB1, APC, AXIN1 and AXIN2 genes (Tate et al. 2019). Although most of the mutations promote stabilization of β-catenin and pathway hyperactivation, several studies have reported that an additional epigenetic inhibition of WNT antagonists is important for cancer formation (Anastas and Moon 2013; Suzuki et al. 2008). In different tumor entities, an epigenetic silencing of various WNT antagonist, such as WIFs, DKKs and SFRPs, correlates with a poor prognosis or high-grade cancer (Kardum et al. 2017; Lin et al. 2017; Davaadorj et al. 2016). Interestingly, a restoration of the WNT antagonist expression attenuated tumor growth (Shih et al. 2007; Gumz et al. 2007). In the present study, we addressed the role of the WNT inhibitory factor SFRP1 in HB and pediatric HCC, since its functional relevance has not been specified, yet. By determining the endogenous gene expression and DNA methylation status of the WNT antagonists APC, DKK1, SFRP1 and WIF1 in four HB cell lines, we detected a heterogeneous pattern. Although all four genes were methylated in HB cells, with the exception of DKK1 in HuH-6 cells, only APC and SFRP1 revealed a concomitant transcriptional repression. Moreover, a treatment of the tumor cells with 5-aza solely restored SFRP1 expression in all four cell lines, indicating that DNA methylation is responsible for the SFRP1 suppression in HB cell lines. We detected no involvement of histone acetylation on SFRP1 regulatory gene sites. Nevertheless, besides DNA methylation, histone modifications might control the expression of other WNT antagonist. Based on these results, we focused our functional analyses on SFRP1 and overexpressed the gene in the HB cell lines HuH6, HepT1 and HepG2. In line with studies in HCC and other tumor entities, a re-expression of SFRP1 inhibited tumor cell growth, colony formation and migration of HB tumor cells (Shih et al. 2007; Kaur et al. 2012; Wang et al. 2018). In contrast to Shih et al. (2007), our data demonstrated that cell lines which possess β-catenin deletion mutations, such as HepT1 and HepG2 cells, also exhibited canonical WNT inhibition. Interestingly, SFRP1 re-expression diminished the canonical WNT/β-catenin signaling activity and we detected a downregulation of the WNT target genes MYC and CCND1, although the HB cell lines carry beta-catenin mutations. Moreover, we uncovered that the overall level and cellular localization of β-catenin is not changed after restored SFRP1 expression. Based on these results, we suppose that SFRP1 expression abolishes β-catenin-driven transcription activity. In accordance, it was shown that the SFRP1-mediated inhibition of the WNT pathway was independent from wildtype or mutant β-catenin in colorectal cancer cells (Suzuki et al. 2004). Hence, the epigenetic silencing of the Wnt antagonists SFRP1 is an important mechanism in pediatric liver carcinogenesis, promoting WNT signaling-mediated oncogenic transformation.
Notably, in our pediatric liver cancer cohort, we detected a reduced SFRP1 gene expression in around 62% (28/45) of cases. To our knowledge, we show for the first time that low SFRP1 expression correlated significantly with a worse classification in the PRETEXT stratification system and to β-catenin mutations, indicating that SFRP1 gene silencing in β-catenin mutant cancer leads to a more aggressive cancer growth. SFRP1 repression might potentiate the oncogenic function of the WNT signaling pathway, particularly in β-catenin mutant cancers. Thus, a restoration of the SFRP1 expression through epigenetic drugs or the natural compound flavonoid epigallocatechin-3-gallate (EGCG), as it was shown in our recent study, could be a new therapeutic option for β-catenin mutant pediatric liver cancers (Godeke et al. 2013). In contrast to other studies, the reduced SFRP1 gene expression in our HB patient cohort was not associated to the DNA methylation status (Vincent and Postovit 2017; Kaur et al. 2012). Several patient samples showed a SFRP1 downregulation without a concomitant methylation of the promoter. With the help of the MSP, we determined the methylation status of only a few cytosines in the CpG island, thus we cannot exclude the possibility that adjacent cytosines might be methylated, which would result in gene silencing (Hernandez et al. 2013). Moreover, recent studies proposed a microRNA-dependent inhibition of SFRP1 in different cancer entities, which may also occur in pediatric liver cancer (Ba et al. 2017; Li et al. 2017). Overall, we identified promoter methylation of SFRP1 in 40% (18/45) of the analyzed cases. Importantly, the SFRP1 methylation correlated with HCC-like pediatric liver tumors, with a higher age at diagnosis and with TERT mutations. It needs to be taken into consideration that these clinical features are dependent on each other, since the tumor manifests at older ages in the HCC/TLCT group (Tomlinson and Kappler 2012), which is furthermore characterized by frequent TERT mutations (Eichenmuller et al. 2014). Although in several tumor entities, SFRP1 expression or methylation was proposed as a prognostic biomarker (Davaadorj et al. 2016; Zheng et al. 2015; Atschekzei et al. 2012), this was not the case for our cohort of pediatric liver cancers.

Conclusion

In conclusion, we delineated an important role for the epigenetic silencing of SFRP1 in pediatric liver cancer cell lines and patient samples. Our data demonstrated that β-catenin mutant pediatric liver cancers are accompanied by a suppression of the WNT antagonist SFRP1, which promotes malignant tumor cell characteristics. SFRP1 methylation was highly associated to advanced pediatric liver tumors, with HCC-like features and TERT mutations. However, further studies are needed to clarify whether SFRP1 gene expression or methylation could serve as a potential prognostic or diagnostic biomarker for HB or pediatric HCC.

Acknowledgements

Open Access funding provided by Projekt DEAL. We thank Maria del Socorro Escobar Lopez, Nicole Stadler, Fatemeh Promoli for technical assistance. We thank Dr. Takayuki Fukui (University of Nagoya, Japan) for kindly providing the pSFRP1 expression vector and Dr. Torsten Pietsch (University of Bonn, Germany) for the HepT1 and Hep-T3 cell lines.

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interests.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.
Not applicable.
Each patient gave written informed consent and the study protocol was approved by the Committee of Ethics, LMU Munich, Germany.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
Zurück zum Zitat Aguilera O, Fraga MF, Ballestar E, Paz MF, Herranz M, Espada J et al (2006) Epigenetic inactivation of the Wnt antagonist DICKKOPF-1 (DKK-1) gene in human colorectal cancer. Oncogene 25(29):4116–4121PubMed Aguilera O, Fraga MF, Ballestar E, Paz MF, Herranz M, Espada J et al (2006) Epigenetic inactivation of the Wnt antagonist DICKKOPF-1 (DKK-1) gene in human colorectal cancer. Oncogene 25(29):4116–4121PubMed
Zurück zum Zitat Anastas JN, Moon RT (2013) WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 13(1):11–26PubMed Anastas JN, Moon RT (2013) WNT signalling pathways as therapeutic targets in cancer. Nat Rev Cancer 13(1):11–26PubMed
Zurück zum Zitat Atschekzei F, Hennenlotter J, Janisch S, Grosshennig A, Trankenschuh W, Waalkes S et al (2012) SFRP1 CpG Island methylation locus is associated with renal cell cancer susceptibility and disease recurrence. Epigenetics 7(5):447–457PubMed Atschekzei F, Hennenlotter J, Janisch S, Grosshennig A, Trankenschuh W, Waalkes S et al (2012) SFRP1 CpG Island methylation locus is associated with renal cell cancer susceptibility and disease recurrence. Epigenetics 7(5):447–457PubMed
Zurück zum Zitat Ba S, Xuan Y, Long ZW, Chen HY, Zheng SS (2017) MicroRNA-27a promotes the proliferation and invasiveness of colon cancer cells by targeting SFRP1 through the Wnt/β-catenin signaling pathway. Cell Physiol Biochem 42(5):1920–1933PubMed Ba S, Xuan Y, Long ZW, Chen HY, Zheng SS (2017) MicroRNA-27a promotes the proliferation and invasiveness of colon cancer cells by targeting SFRP1 through the Wnt/β-catenin signaling pathway. Cell Physiol Biochem 42(5):1920–1933PubMed
Zurück zum Zitat Bell D, Ranganathan S, Tao J, Monga SP (2017) Novel Advances in understanding of molecular pathogenesis of hepatoblastoma: a Wnt/β-catenin perspective. Gene Exp 17(2):141–154 Bell D, Ranganathan S, Tao J, Monga SP (2017) Novel Advances in understanding of molecular pathogenesis of hepatoblastoma: a Wnt/β-catenin perspective. Gene Exp 17(2):141–154
Zurück zum Zitat Benitz S, Straub T, Mahajan UM, Mutter J, Czemmel S, Unruh T et al (2019) Ring1b-dependent epigenetic remodelling is an essential prerequisite for pancreatic carcinogenesis. Gut 68(11):2007–2018PubMed Benitz S, Straub T, Mahajan UM, Mutter J, Czemmel S, Unruh T et al (2019) Ring1b-dependent epigenetic remodelling is an essential prerequisite for pancreatic carcinogenesis. Gut 68(11):2007–2018PubMed
Zurück zum Zitat Cairo S, Armengol C, De Reynies A, Wei Y, Thomas E, Renard CA et al (2008) Hepatic stem-like phenotype and interplay of Wnt/beta-catenin and Myc signaling in aggressive childhood liver cancer. Cancer Cell 14(6):471–484PubMed Cairo S, Armengol C, De Reynies A, Wei Y, Thomas E, Renard CA et al (2008) Hepatic stem-like phenotype and interplay of Wnt/beta-catenin and Myc signaling in aggressive childhood liver cancer. Cancer Cell 14(6):471–484PubMed
Zurück zum Zitat Czauderna P, Garnier H (2018) Hepatoblastoma: current understanding, recent advances, and controversies. F1000Res 7:53PubMedPubMedCentral Czauderna P, Garnier H (2018) Hepatoblastoma: current understanding, recent advances, and controversies. F1000Res 7:53PubMedPubMedCentral
Zurück zum Zitat Czauderna P, Haeberle B, Hiyama E, Rangaswami A, Krailo M, Maibach R et al (2016) The Children’s Hepatic tumors International Collaboration (CHIC): novel global rare tumor database yields new prognostic factors in hepatoblastoma and becomes a research model. Eur J Cancer 52:92–101PubMed Czauderna P, Haeberle B, Hiyama E, Rangaswami A, Krailo M, Maibach R et al (2016) The Children’s Hepatic tumors International Collaboration (CHIC): novel global rare tumor database yields new prognostic factors in hepatoblastoma and becomes a research model. Eur J Cancer 52:92–101PubMed
Zurück zum Zitat Davaadorj M, Imura S, Saito YU, Morine Y, Ikemoto T, Yamada S et al (2016) Loss of SFRP1 expression is associated with poor prognosis in hepatocellular carcinoma. Anticancer Res 36(2):659–664PubMed Davaadorj M, Imura S, Saito YU, Morine Y, Ikemoto T, Yamada S et al (2016) Loss of SFRP1 expression is associated with poor prognosis in hepatocellular carcinoma. Anticancer Res 36(2):659–664PubMed
Zurück zum Zitat Eichenmuller M, Gruner I, Hagl B, Haberle B, Muller-Hocker J, von Schweinitz D et al (2009) Blocking the hedgehog pathway inhibits hepatoblastoma growth. Hepatology 49(2):482–490PubMed Eichenmuller M, Gruner I, Hagl B, Haberle B, Muller-Hocker J, von Schweinitz D et al (2009) Blocking the hedgehog pathway inhibits hepatoblastoma growth. Hepatology 49(2):482–490PubMed
Zurück zum Zitat Eichenmuller M, Trippel F, Kreuder M, Beck A, Schwarzmayr T, Haberle B et al (2014) The genomic landscape of hepatoblastoma and their progenies with HCC-like features. J Hepatol 61(6):1312–1320PubMed Eichenmuller M, Trippel F, Kreuder M, Beck A, Schwarzmayr T, Haberle B et al (2014) The genomic landscape of hepatoblastoma and their progenies with HCC-like features. J Hepatol 61(6):1312–1320PubMed
Zurück zum Zitat Esteller M, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA et al (2000) Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res 60(16):4366–4371PubMed Esteller M, Sparks A, Toyota M, Sanchez-Cespedes M, Capella G, Peinado MA et al (2000) Analysis of adenomatous polyposis coli promoter hypermethylation in human cancer. Cancer Res 60(16):4366–4371PubMed
Zurück zum Zitat Fukui T, Kondo M, Ito G, Maeda O, Sato N, Yoshioka H et al (2005) Transcriptional silencing of secreted frizzled related protein 1 (SFRP 1) by promoter hypermethylation in non-small-cell lung cancer. Oncogene 24(41):6323–6327PubMed Fukui T, Kondo M, Ito G, Maeda O, Sato N, Yoshioka H et al (2005) Transcriptional silencing of secreted frizzled related protein 1 (SFRP 1) by promoter hypermethylation in non-small-cell lung cancer. Oncogene 24(41):6323–6327PubMed
Zurück zum Zitat Godeke J, Maier S, Eichenmuller M, Muller-Hocker J, von Schweinitz D, Kappler R (2013) Epigallocatechin-3-gallate inhibits hepatoblastoma growth by reactivating the Wnt inhibitor SFRP1. Nutr Cancer 65(8):1200–1207PubMed Godeke J, Maier S, Eichenmuller M, Muller-Hocker J, von Schweinitz D, Kappler R (2013) Epigallocatechin-3-gallate inhibits hepatoblastoma growth by reactivating the Wnt inhibitor SFRP1. Nutr Cancer 65(8):1200–1207PubMed
Zurück zum Zitat Gumz ML, Zou H, Kreinest PA, Childs AC, Belmonte LS, LeGrand SN et al (2007) Secreted frizzled-related protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res 13(16):4740–4749PubMed Gumz ML, Zou H, Kreinest PA, Childs AC, Belmonte LS, LeGrand SN et al (2007) Secreted frizzled-related protein 1 loss contributes to tumor phenotype of clear cell renal cell carcinoma. Clin Cancer Res 13(16):4740–4749PubMed
Zurück zum Zitat Hernandez HG, Tse MY, Pang SC, Arboleda H, Forero DA (2013) Optimizing methodologies for PCR-based DNA methylation analysis. Biotechniques 55(4):181–197PubMed Hernandez HG, Tse MY, Pang SC, Arboleda H, Forero DA (2013) Optimizing methodologies for PCR-based DNA methylation analysis. Biotechniques 55(4):181–197PubMed
Zurück zum Zitat Huang J, Zhang YL, Teng XM, Lin Y, Zheng DL, Yang PY et al (2007) Down-regulation of SFRP1 as a putative tumor suppressor gene can contribute to human hepatocellular carcinoma. BMC Cancer 7:126PubMedPubMedCentral Huang J, Zhang YL, Teng XM, Lin Y, Zheng DL, Yang PY et al (2007) Down-regulation of SFRP1 as a putative tumor suppressor gene can contribute to human hepatocellular carcinoma. BMC Cancer 7:126PubMedPubMedCentral
Zurück zum Zitat Kardum V, Karin V, Glibo M, Skrtic A, Martic TN, Ibisevic N et al (2017) Methylation-associated silencing of SFRP1 gene in high-grade serous ovarian carcinomas. Ann Diagn Pathol 31:45–49PubMed Kardum V, Karin V, Glibo M, Skrtic A, Martic TN, Ibisevic N et al (2017) Methylation-associated silencing of SFRP1 gene in high-grade serous ovarian carcinomas. Ann Diagn Pathol 31:45–49PubMed
Zurück zum Zitat Kaur P, Mani S, Cros MP, Scoazec JY, Chemin I, Hainaut P et al (2012) Epigenetic silencing of sFRP1 activates the canonical Wnt pathway and contributes to increased cell growth and proliferation in hepatocellular carcinoma. Tumour Biol 33(2):325–336PubMed Kaur P, Mani S, Cros MP, Scoazec JY, Chemin I, Hainaut P et al (2012) Epigenetic silencing of sFRP1 activates the canonical Wnt pathway and contributes to increased cell growth and proliferation in hepatocellular carcinoma. Tumour Biol 33(2):325–336PubMed
Zurück zum Zitat Kawano Y, Kypta R (2003) Secreted antagonists of the Wnt signalling pathway. J Cell Sci 116(Pt 13):2627–2634PubMed Kawano Y, Kypta R (2003) Secreted antagonists of the Wnt signalling pathway. J Cell Sci 116(Pt 13):2627–2634PubMed
Zurück zum Zitat Kremer N, Walther AE, Tiao GM (2014) Management of hepatoblastoma: an update. Curr Opin Pediatr 26(3):362–369PubMed Kremer N, Walther AE, Tiao GM (2014) Management of hepatoblastoma: an update. Curr Opin Pediatr 26(3):362–369PubMed
Zurück zum Zitat Li H, Yang T, Shang D, Sun Z (2017) miR-1254 promotes lung cancer cell proliferation by targeting SFRP1. Biomed Pharmacother 92:913–918PubMed Li H, Yang T, Shang D, Sun Z (2017) miR-1254 promotes lung cancer cell proliferation by targeting SFRP1. Biomed Pharmacother 92:913–918PubMed
Zurück zum Zitat Lin B, Hong H, Jiang X, Li C, Zhu S, Tang N et al (2017) WNT inhibitory factor 1 promoter hypermethylation is an early event during gallbladder cancer tumorigenesis that predicts poor survival. Gene 622:42–49PubMed Lin B, Hong H, Jiang X, Li C, Zhu S, Tang N et al (2017) WNT inhibitory factor 1 promoter hypermethylation is an early event during gallbladder cancer tumorigenesis that predicts poor survival. Gene 622:42–49PubMed
Zurück zum Zitat Lo PK, Mehrotra J, D'Costa A, Fackler MJ, Garrett-Mayer E, Argani P et al (2006) Epigenetic suppression of secreted frizzled related protein 1 (SFRP1) expression in human breast cancer. Cancer Biol Ther 5(3):281–286PubMed Lo PK, Mehrotra J, D'Costa A, Fackler MJ, Garrett-Mayer E, Argani P et al (2006) Epigenetic suppression of secreted frizzled related protein 1 (SFRP1) expression in human breast cancer. Cancer Biol Ther 5(3):281–286PubMed
Zurück zum Zitat Lodygin D, Epanchintsev A, Menssen A, Diebold J, Hermeking H (2005) Functional epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res 65(10):4218–4227PubMed Lodygin D, Epanchintsev A, Menssen A, Diebold J, Hermeking H (2005) Functional epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res 65(10):4218–4227PubMed
Zurück zum Zitat Ng K, Mogul DB (2018) Pediatric liver tumors. Clin Liver Dis 22(4):753–772PubMed Ng K, Mogul DB (2018) Pediatric liver tumors. Clin Liver Dis 22(4):753–772PubMed
Zurück zum Zitat Perugorria MJ, Olaizola P, Labiano I, Esparza-Baquer A, Marzioni M, Marin JJG et al (2019) Wnt-beta-catenin signalling in liver development, health and disease. Nat Rev Gastroenterol Hepatol 16(2):121–136PubMed Perugorria MJ, Olaizola P, Labiano I, Esparza-Baquer A, Marzioni M, Marin JJG et al (2019) Wnt-beta-catenin signalling in liver development, health and disease. Nat Rev Gastroenterol Hepatol 16(2):121–136PubMed
Zurück zum Zitat Russell JO, Monga SP (2018) Wnt/β-catenin signaling in liver development, homeostasis, and pathobiology. Annu Rev Pathol 13:351–378PubMed Russell JO, Monga SP (2018) Wnt/β-catenin signaling in liver development, homeostasis, and pathobiology. Annu Rev Pathol 13:351–378PubMed
Zurück zum Zitat Shih YL, Shyu RY, Hsieh CB, Lai HC, Liu KY, Chu TY et al (2006) Promoter methylation of the secreted frizzled-related protein 1 gene SFRP1 is frequent in hepatocellular carcinoma. Cancer 107(3):579–590PubMed Shih YL, Shyu RY, Hsieh CB, Lai HC, Liu KY, Chu TY et al (2006) Promoter methylation of the secreted frizzled-related protein 1 gene SFRP1 is frequent in hepatocellular carcinoma. Cancer 107(3):579–590PubMed
Zurück zum Zitat Shih YL, Hsieh CB, Lai HC, Yan MD, Hsieh TY, Chao YC et al (2007) SFRP1 suppressed hepatoma cells growth through Wnt canonical signaling pathway. Int J Cancer 121(5):1028–1035PubMed Shih YL, Hsieh CB, Lai HC, Yan MD, Hsieh TY, Chao YC et al (2007) SFRP1 suppressed hepatoma cells growth through Wnt canonical signaling pathway. Int J Cancer 121(5):1028–1035PubMed
Zurück zum Zitat Suzuki H, Watkins DN, Jair KW, Schuebel KE, Markowitz SD, Chen WD et al (2004) Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet 36(4):417–422PubMed Suzuki H, Watkins DN, Jair KW, Schuebel KE, Markowitz SD, Chen WD et al (2004) Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat Genet 36(4):417–422PubMed
Zurück zum Zitat Suzuki H, Toyota M, Carraway H, Gabrielson E, Ohmura T, Fujikane T et al (2008) Frequent epigenetic inactivation of Wnt antagonist genes in breast cancer. Br J Cancer 98(6):1147–1156PubMedPubMedCentral Suzuki H, Toyota M, Carraway H, Gabrielson E, Ohmura T, Fujikane T et al (2008) Frequent epigenetic inactivation of Wnt antagonist genes in breast cancer. Br J Cancer 98(6):1147–1156PubMedPubMedCentral
Zurück zum Zitat Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N et al (2019) COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res 47(D1):D941–D947PubMed Tate JG, Bamford S, Jubb HC, Sondka Z, Beare DM, Bindal N et al (2019) COSMIC: the catalogue of somatic mutations in cancer. Nucleic Acids Res 47(D1):D941–D947PubMed
Zurück zum Zitat Tomlinson GE, Kappler R (2012) Genetics and epigenetics of hepatoblastoma. Pediatr Blood Cancer 59(5):785–792PubMed Tomlinson GE, Kappler R (2012) Genetics and epigenetics of hepatoblastoma. Pediatr Blood Cancer 59(5):785–792PubMed
Zurück zum Zitat Towbin AJ, Meyers RL, Woodley H, Miyazaki O, Weldon CB, Morland B et al (2018) 2017 PRETEXT: radiologic staging system for primary hepatic malignancies of childhood revised for the Paediatric Hepatic International Tumour Trial (PHITT). Pediatr Radiol 48(4):536–554PubMed Towbin AJ, Meyers RL, Woodley H, Miyazaki O, Weldon CB, Morland B et al (2018) 2017 PRETEXT: radiologic staging system for primary hepatic malignancies of childhood revised for the Paediatric Hepatic International Tumour Trial (PHITT). Pediatr Radiol 48(4):536–554PubMed
Zurück zum Zitat Tulla M, Berthold F, Graf N, Rutkowski S, von Schweinitz D, Spix C et al (2015) Incidence, trends, and survival of children with embryonal tumors. Pediatrics 136(3):e623–e632PubMed Tulla M, Berthold F, Graf N, Rutkowski S, von Schweinitz D, Spix C et al (2015) Incidence, trends, and survival of children with embryonal tumors. Pediatrics 136(3):e623–e632PubMed
Zurück zum Zitat Vincent KM, Postovit LM (2017) A pan-cancer analysis of secreted Frizzled-related proteins: re-examining their proposed tumour suppressive function. Sci Rep 7:42719PubMedPubMedCentral Vincent KM, Postovit LM (2017) A pan-cancer analysis of secreted Frizzled-related proteins: re-examining their proposed tumour suppressive function. Sci Rep 7:42719PubMedPubMedCentral
Zurück zum Zitat Wang Z, Li R, He Y, Huang S (2018) Effects of secreted frizzled-related protein 1 on proliferation, migration, invasion, and apoptosis of colorectal cancer cells. Cancer Cell Int 18:48PubMedPubMedCentral Wang Z, Li R, He Y, Huang S (2018) Effects of secreted frizzled-related protein 1 on proliferation, migration, invasion, and apoptosis of colorectal cancer cells. Cancer Cell Int 18:48PubMedPubMedCentral
Zurück zum Zitat Wirths O, Waha A, Weggen S, Schirmacher P, Kuhne T, Goodyer CG et al (2003) Overexpression of human Dickkopf-1, an antagonist of wingless/WNT signaling, in human hepatoblastomas and Wilms’ tumors. Lab Invest 83(3):429–434PubMed Wirths O, Waha A, Weggen S, Schirmacher P, Kuhne T, Goodyer CG et al (2003) Overexpression of human Dickkopf-1, an antagonist of wingless/WNT signaling, in human hepatoblastomas and Wilms’ tumors. Lab Invest 83(3):429–434PubMed
Zurück zum Zitat Zheng L, Sun D, Fan W, Zhang Z, Li Q, Jiang T (2015) Diagnostic value of SFRP1 as a favorable predictive and prognostic biomarker in patients with prostate cancer. PLoS ONE 10(2):e0118276PubMedPubMedCentral Zheng L, Sun D, Fan W, Zhang Z, Li Q, Jiang T (2015) Diagnostic value of SFRP1 as a favorable predictive and prognostic biomarker in patients with prostate cancer. PLoS ONE 10(2):e0118276PubMedPubMedCentral
Zurück zum Zitat Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM (2015) Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology 149(5):1226–1239 (e4) PubMed Zucman-Rossi J, Villanueva A, Nault JC, Llovet JM (2015) Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology 149(5):1226–1239 (e4) PubMed
Metadaten
Titel
Downregulation of SFRP1 is a protumorigenic event in hepatoblastoma and correlates with beta-catenin mutations
verfasst von
Ivonne Regel
Melanie Eichenmüller
Ujjwal Mukund Mahajan
Beate Hagl
Simone Benitz
Beate Häberle
Christian Vokuhl
Dietrich von Schweinitz
Roland Kappler
Publikationsdatum
18.03.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 5/2020
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-020-03182-1

Weitere Artikel der Ausgabe 5/2020

Journal of Cancer Research and Clinical Oncology 5/2020 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.