Skip to main content
Erschienen in: BMC Cancer 1/2021

Open Access 01.12.2021 | Research

METTL14 gene polymorphisms decrease Wilms tumor susceptibility in Chinese children

verfasst von: Zhenjian Zhuo, Rui-Xi Hua, Huizhu Zhang, Huiran Lin, Wen Fu, Jinhong Zhu, Jiwen Cheng, Jiao Zhang, Suhong Li, Haixia Zhou, Huimin Xia, Guochang Liu, Wei Jia, Jing He

Erschienen in: BMC Cancer | Ausgabe 1/2021

Abstract

Background

Wilms tumor is a highly heritable malignancy. Aberrant METTL14, a critical component of N6-methyladenosine (m6A) methyltransferase, is involved in carcinogenesis. The association between genetic variants in the METTL14 gene and Wilms tumor susceptibility remains to be fully elucidated. We aimed to assess whether variants within this gene are implicated in Wilms tumor susceptibility.

Methods

A total of 403 patients and 1198 controls were analyzed. METTL14 genotypes were assessed by TaqMan genotyping assay.

Result

Among the five SNPs analyzed, rs1064034 T > A and rs298982 G > A exhibited a significant association with decreased susceptibility to Wilms tumor. Moreover, the joint analysis revealed that the combination of five protective genotypes exerted significantly more protective effects against Wilms tumor than 0–4 protective genotypes with an OR of 0.69. The stratified analysis further identified the protective effect of rs1064034 T > A, rs298982 G > A, and combined five protective genotypes in specific subgroups. The above significant associations were further validated by haplotype analysis and false-positive report probability analysis. Preliminary mechanism exploration indicated that rs1064034 T > A and rs298982 G > A are correlated with the expression and splicing event of their surrounding genes.

Conclusions

Collectively, our results suggest that METTL14 gene SNPs may be genetic modifiers for the development of Wilms tumor.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-021-09019-5.
Zhenjian Zhuo, Rui-Xi Hua and Huizhu Zhang contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
m6A
N6-methyladenosine
HWE
Hardy-Weinberg equilibrium
ORs
Odds ratios
CIs
Confidence intervals
FPRP
False-positive report probability analysis
eQTL
Expression quantitative trait loci
sQTLs
Splicing quantitative trait loci
GTEx
Genotype-Tissue Expression
AS
Alternative splicing

Introduction

Wilms tumor, also known as nephroblastoma, is the most common pediatric kidney cancer [1]. It accounts for over 90% of all the diagnosed kidney tumors in children [2]. The incidence rate of Wilms’ tumor varies geographically [3, 4]. The prevalence of Wilms tumor is about 7 cases per million children in the United States. Wilms tumor is also one of the most common renal tumors in children in China, with an incidence rate of ~ 3.3 per million. Wilms tumors are frequently diagnosed in young children with an average age of 2–3 years [5]. At present, long-term overall survival for the localized Wilms tumors exceeds 90% due to the improved risk stratification-adapted treatment [6]. However, nearly 20% of Wilms tumors are classified into high-risk subtype with frequent metastasis. Patients with high-risk tumors still subject to suboptimal outcomes [79]. Chronic health conditions secondary to intensified therapeutic regimens impact nearly 25% of Wilms tumor survivors [10].
The genetics of Wilms tumor tumorigenesis is complex, with multiple oncogenic drivers identified over the years. The currently known repertoire of oncogenic Wilms tumor driver alterations includes mutations in the WT1, CTNNB1, TP53, AMER1, as well as an abnormality of 11p15 methylation [1115]. Apart from these, genetic association analyses in case-control studies also unveiled some Wilms tumor susceptibility loci [1619]. Nevertheless, the well-established risk factors for Wilms tumor probably are only the tip of the iceberg. So far, all the known gene mutations can only explain less than 50% of Wilms tumor. Therefore, it is imperative to identify more causative variants to improve the understanding of the genetic susceptibility to Wilms tumor. In addition, detailed genetic information leads to new druggable targets, facilitating the development of more effective treatments for Wilms tumor.
N6-methyladenosine (m6A) is the most common internal chemical modification on eukaryotic mRNA [20]. m6A is mainly involved in the regulation of splicing, subcellular localization, translation, stability, and degradation of mRNA. m6A modulators are mainly classified into methyltransferase (writer), demethylase (eraser), and binding protein (reader). Methyltransferases include METTL3, METTL14, and WTAP, which mainly mediate m6A methylation of mRNA adenylate. Demethylases, consisting of FTO and ALKBH5, mainly remove m6A modification installed on RNA. Binding proteins include YTHDF1/2/3, YTHDC1/2, IGF2BP1/2/3, and eIF3, which are responsible for recognizing bases modified by m6A and regulating downstream pathways [21, 22]. The m6A modulator proteins play an important role in the occurrence and development of a variety of tumors [2325]. However, research on the expression and function of m6A modulator genes in Wilms tumor has not yet been reported. The scarcity of investigation prompted us to contribute to our current report on associations between genetic variability of METTL14 and the risk of Wilms tumor. To this end, a total of five common SNPs in the METTL14 gene were genotyped and tested for their association with Wilms tumor susceptibility.

Methods

Sample selection

The study was carried out based on the principles of the Declaration of Helsinki. Approval of the study protocol was obtained from the institutional review board of Guangzhou Women and Children’s Medical Center (Ethics Approval No: 202016600). Eligible cases were all children newly diagnosed with a histologically confirmed Wilms tumor. Controls, recruited from the same hospital, were healthy volunteers of Chinese origin, without family history of Wilms tumor. Written informed consent was signed by all subjects’ guardians. All the subjects were enrolled from March 2001 to March 2018 and were genetically unrelated ethnic Han Chinese from China. A total of 414 cases diagnosed with Wilms tumor and 1199 hospital-based controls were included. They were enrolled from five hospitals (Guangzhou Women and Children’s Medical Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, The First Affiliated Hospital of Zhengzhou University, Second Affiliated Hospital of Xi’an Jiao Tong University, and Shanxi Provincial Children’s Hospital) in five different cities of China. Detailed information was previously reported [26, 27].

Polymorphism selection and genotyping

The selection of the five potentially functional METTL14 gene SNPs (rs1064034 T > A, rs298982 G > A, rs62328061 A > G, rs9884978 G > A, and rs4834698 T > C) was described in detail in our previous studies [2830]. Genomic DNA from each sample was extracted from peripheral blood. Genotypes were determined using the TaqMan method. Replicate samples (10% of the samples) were picked out of all genotyping batches, and the concordance levels for blind duplicate samples were 100% for all SNPs assayed.

Statistical analysis

SNP genotypes were tested for consistency with Hardy-Weinberg equilibrium (HWE) within the control sample using a Goodness-of-fit χ2 test. Differences between cases and controls in the distribution of demographic and clinical variables were checked using a two-sided χ2 test. Adjusted odds ratios (ORs) with 95% confidence intervals (CIs) and two-sided P-values were calculated using unconditional logistic regression to estimate the relative risk associated with each genotype. Associations were further estimated in the groups stratified by age, gender, and clinical stages. Haplotype frequency distributions were deduced from observed genotypes using logistic regression analyses [31, 32]. False-positive report probability (FPRP) analysis was applied to assess noteworthy associations with detailed methods presented elsewhere [33, 34]. We performed expression quantitative trait loci (eQTL) and splicing quantitative trait loci (sQTLs) analyses through the Genotype-Tissue Expression (GTEx) project (http://​www.​gtexportal.​org/​) to evaluate the correlations between genotypes of candidate SNPs and genes expression as well as alternative splicing (AS) events of genes [35]. A probability value (P value) less than 0.05 was considered significant. All statistical analyses were performed using SAS version 9.1 software (SAS Institute, Inc., Cary, North Carolina).

Results

Effect of METTL14 gene SNPs on Wilms tumor risk

Clinical characteristics of the participants were depicted in our previous study (Table S1) [27]. Here, we successfully genotyped the five METTL14 gene SNPs (rs1064034 T > A, rs298982 G > A, rs62328061 A > G, rs9884978 G > A, and rs4834698 T > C) in 403 cases and 1198 controls, out of 414 cases and 1199 controls samples. The correlation between these SNPs and Wilms tumor risk is shown in Table 1. All these SNPs followed Hardy-Weinberg equilibrium (HWE) in controls (HWE P > 0.05). The rs1064034 variant alleles were remarkably associated with reduced risk of Wilms tumor (TA vs. TT: adjusted OR = 0.78, 95% CI = 0.61–0.99, P = 0.041; TA/AA vs. TT: adjusted OR = 0.83, 95% CI = 0.70–0.995, P = 0.044). Similar association was found for the rs298982 (GA/AA vs. GG: adjusted OR = 0.69, 95% CI = 0.53–0.91, P = 0.009). We then defined rs1064034 TA/AA, rs298982 GA/AA, rs62328061 AG/AA, rs9884978 GA/GG, and rs4834698 TT/TC as protective genotypes based on their ORs. Participants with 5 protective genotypes showed a 0.69-fold decrease in the risk of developing Wilms tumor when compared with those with 0–4 protective genotypes (95% CI = 0.52–0.91, P = 0.008).
Table 1
Association between METTL14 gene polymorphisms and Wilms tumor susceptibility
Genotype
Cases (N = 403)
Controls (N = 1198)
Pa
Crude OR (95% CI)
P
Adjusted OR (95% CI) b
Pb
rs1064034 T > A (HWE = 0.715)
 TT
216 (53.60)
564 (47.08)
 
1.00
 
1.00
 
 TA
152 (37.72)
512 (42.74)
 
0.78 (0.61–0.99)
0.037
0.78 (0.61–0.99)
0.041
 AA
35 (8.68)
122 (10.18)
 
0.75 (0.50–1.13)
0.164
0.76 (0.51–1.15)
0.198
 Additive
  
0.035
0.83 (0.70–0.99)
0.035
0.83 (0.70–0.995)
0.044
 Dominant
187 (46.40)
634 (52.92)
0.024
0.77 (0.61–0.97)
0.024
0.78 (0.62–0.97)
0.029
 Recessive
368 (91.32)
1076 (89.82)
0.382
0.84 (0.57–1.24)
0.382
0.86 (0.58–1.27)
0.438
rs298982 G > A (HWE = 0.155)
 GG
321 (79.65)
873 (72.87)
 
1.00
 
1.00
 
 GA
66 (16.38)
292 (24.37)
 
0.62 (0.46–0.83)
0.001
0.62 (0.46–0.84)
0.002
 AA
16 (3.97)
33 (2.75)
 
1.32 (0.72–2.43)
0.375
1.32 (0.72–2.43)
0.373
 Additive
  
0.061
0.80 (0.64–1.01)
0.061
0.81 (0.64–1.02)
0.071
 Dominant
82 (20.35)
325 (27.13)
0.007
0.69 (0.52–0.90)
0.007
0.69 (0.53–0.91)
0.009
 Recessive
387 (96.03)
1165 (97.25)
0.220
1.46 (0.80–2.68)
0.223
1.46 (0.79–2.68)
0.225
rs62328061 A > G (HWE = 0.819)
 AA
281 (69.73)
830 (69.28)
 
1.00
 
1.00
 
 AG
109 (27.05)
333 (27.80)
 
0.97 (0.75–1.25)
0.796
0.97 (0.75–1.25)
0.812
 GG
13 (3.23)
35 (2.92)
 
1.10 (0.57–2.10)
0.780
1.12 (0.58–2.15)
0.736
 Additive
  
0.963
1.00 (0.81–1.23)
0.963
1.00 (0.81–1.24)
0.998
 Dominant
122 (30.27)
368 (30.72)
0.867
0.98 (0.77–1.25)
0.867
0.98 (0.77–1.26)
0.894
 Recessive
390 (96.77)
1163 (97.08)
0.757
1.11 (0.58–2.12)
0.757
1.13 (0.59–2.16)
0.714
rs9884978 G > A (HWE = 0.412)
 GG
252 (62.53)
758 (63.27)
 
1.00
 
1.00
 
 GA
131 (32.51)
384 (32.05)
 
1.03 (0.80–1.31)
0.836
1.03 (0.81–1.31)
0.826
 AA
20 (4.96)
56 (4.67)
 
1.07 (0.63–1.83)
0.791
1.06 (0.62–1.80)
0.826
 Additive
  
0.759
1.03 (0.85–1.25)
0.757
1.03 (0.85–1.25)
0.773
 Dominant
151 (37.47)
440 (36.73)
0.790
1.03 (0.82–1.30)
0.789
1.03 (0.82–1.30)
0.791
 Recessive
383 (95.04)
1142 (95.33)
0.814
1.07 (0.63–1.80)
0.814
1.05 (0.62–1.78)
0.851
rs4834698 T > C (HWE = 0.827)
 TT
107 (26.55)
329 (27.46)
 
1.00
 
1.00
 
 TC
193 (47.89)
594 (49.58)
 
1.00 (0.76–1.31)
0.995
0.99 (0.75–1.30)
0.921
 CC
103 (25.56)
275 (22.95)
 
1.15 (0.84–1.58)
0.379
1.14 (0.83–1.56)
0.425
 Additive
  
0.392
1.07 (0.92–1.26)
0.392
1.07 (0.91–1.25)
0.438
 Dominant
296 (73.45)
869 (72.54)
0.722
1.05 (0.81–1.35)
0.724
1.03 (0.80–1.34)
0.798
 Recessive
300 (74.44)
923 (77.05)
0.287
1.15 (0.89–1.50)
0.287
1.15 (0.88–1.49)
0.304
Combined effect of protective genotypes c
 0–4
322 (79.90)
875 (73.04)
 
1.00
 
1.00
 
 5
81 (20.10)
323 (26.96)
0.006
0.68 (0.52–0.90)
0.006
0.69 (0.52–0.91)
0.008
OR Odds ratio, CI Confidence interval, HWE Hardy-Weinberg equilibrium
aχ2 test for genotype distributions between Wilms tumor patients and controls
bAdjusted for age and gender
cProtective genotypes were carriers with rs1064034 TA/AA, rs298982 GA/AA, rs62328061 AG/AA, rs9884978 GA/GG and rs4834698 TT/TC

Stratification analysis of significant SNPs

We analyzed the association between the METTL14 gene polymorphisms and susceptibility to Wilms tumor in subgroups separated by age, gender, and clinical stages (Table 2). Further stratification study revealed that the rs1064034 was associated with reduced Wilms tumor risk in groups with age > 18 months, female, and clinical stage IV diseases. Moreover, stronger protective effects was found for the GA/AA genotypes of rs298982 and combined five protective genotypes among children age > 18 months, females, clinical stage I + II tumors, and clinical stage III + IV tumors.
Table 2
Stratification analysis of protective genotypes with Wilms tumor susceptibility
Variables
rs1064034 (cases/controls)
AOR (95% CI) a
Pa
rs298982 (cases/controls)
AOR (95% CI) a
Pa
Combined (cases/controls)
AOR (95% CI) a
Pa
TT
TA/AA
GG
GA/AA
0–4
5
Age, month
  ≤ 18
72/243
66/222
1.00 (0.68–1.47)
0.995
105/356
33/109
1.01 (0.65–1.58)
0.971
106/358
32/107
0.99 (0.63–1.56)
0.967
  > 18
144/321
121/412
0.67 (0.50–0.88)
0.005
216/517
49/216
0.56 (0.39–0.79)
0.001
216/517
49/216
0.56 (0.39–0.79)
0.001
Gender
 Females
109/251
80/270
0.68 (0.49–0.95)
0.025
159/394
30/127
0.59 (0.38–0.91)
0.017
159/396
30/125
0.60 (0.39–0.93)
0.022
 Males
107/313
107/364
0.87 (0.64–1.18)
0.371
162/479
52/198
0.78 (0.55–1.11)
0.172
163/479
51/198
0.76 (0.53–1.09)
0.134
Clinical stages
 I
73/564
64/634
0.81 (0.57–1.15)
0.239
111/873
26/325
0.64 (0.41–1.01)
0.053
111/875
26/323
0.65 (0.42–1.02)
0.060
 II
61/564
52/634
0.77 (0.52–1.14)
0.193
88/873
25/325
0.78 (0.49–1.23)
0.285
88/875
25/323
0.79 (0.49–1.25)
0.305
 III
44/564
48/634
0.94 (0.61–1.44)
0.781
74/873
18/325
0.64 (0.38–1.10)
0.105
74/875
18/323
0.65 (0.38–1.10)
0.111
 IV
28/564
17/634
0.53 (0.29–0.98)
0.043
37/873
8/325
0.58 (0.27–1.26)
0.171
38/875
7/323
0.50 (0.22–1.13)
0.095
 I + II
134/564
116/634
0.79 (0.60–1.04)
0.093
199/873
51/325
0.70 (0.50–0.98)
0.037
199/875
51/323
0.71 (0.51–0.99)
0.043
 III + IV
72/564
65/634
0.79 (0.55–1.12)
0.183
111/873
26/325
0.62 (0.40–0.98)
0.039
112/875
25/323
0.60 (0.38–0.94)
0.026
AOR Adjusted odds ratio, CI Confidence interval
aAdjusted for age and gender, omitting the corresponding factor

METTL14 haplotype analysis

We next evaluated whether the haplotypes of the five METTL14 gene SNPs are linked with Wilms tumor risk (Table 3). When compared to reference haplotype TGAAC, haplotypes AGAGT (P = 0.016), AAGGT (P = 0.010), and AAAGC (P = 0.002) were linked with significantly decreased Wilms tumor risk.
Table 3
The frequency of inferred haplotypes of METTL14 gene based on observed genotypes and their association with the risk of Wilms tumor
Haplotypes a
Cases (n = 806)
Controls (n = 2396)
Crude OR (95% CI)
P
Adjusted OR b (95% CI)
Pb
TGAAC
78 (9.68)
233 (9.72)
1.00
 
1.00
 
TGAAT
41 (5.09)
111 (4.63)
0.88 (0.57–1.34)
0.542
0.87 (0.57–1.33)
0.516
TGAGC
209 (25.93)
550 (22.95)
0.90 (0.68–1.20)
0.468
0.90 (0.68–1.19)
0.464
TGAGT
242 (30.02)
744 (31.05)
0.77 (0.59–1.02)
0.064
0.77 (0.59–1.02)
0.066
TGGAT
4 (0.50)
0 (0.00)
/
/
/
/
TGGGC
5 (0.62)
1 (0.04)
11.85 (1.37–102.72)
0.025
11.15 (1.28–96.76)
0.029
TGGGT
3 (0.37)
1 (0.04)
7.11 (0.73–69.18)
0.091
7.50 (0.77–73.05)
0.083
TAAAT
1 (0.12)
0 (0.00)
/
/
/
/
TAAGC
1 (0.12)
0 (0.00)
/
/
/
/
AGGAT
23 (2.85)
79 (3.30)
0.69 (0.41–1.16)
0.162
0.70 (0.41–1.16)
0.172
AGGGC
65 (8.06)
193 (8.06)
0.80 (0.55–1.15)
0.227
0.80 (0.55–1.15)
0.221
AGGGT
23 (2.85)
69 (2.88)
0.79 (0.47–1.34)
0.380
0.80 (0.47–1.36)
0.417
AGAAC
3 (0.37)
0 (0.00)
/
/
/
/
AGAAT
2 (0.25)
1 (0.04)
4.74 (0.43–52.87)
0.206
5.23 (0.47–58.94)
0.180
AGAGC
1 (0.12)
1 (0.04)
2.37 (0.15–38.27)
0.543
2.46 (0.15–39.70)
0.527
AGAGT
9 (1.12)
55 (2.30)
0.39 (0.19–0.82)
0.012
0.40 (0.19–0.84)
0.016
AAGAC
1 (0.12)
0 (0.00)
/
/
/
/
AAGGC
2 (0.25)
2 (0.08)
2.37 (0.33–17.06)
0.392
2.32 (0.32–16.75)
0.403
AAGGT
9 (1.12)
58 (2.42)
0.37 (0.18–0.77)
0.008
0.38 (0.18–0.80)
0.010
AAAAC
0 (0.00)
2 (0.08)
/
/
/
/
AAAAT
18 (2.23)
70 (2.92)
0.61 (0.35–1.08)
0.088
0.62 (0.35–1.09)
0.096
AAAGC
34 (4.22)
162 (6.76)
0.50 (0.32–0.77)
0.002
0.50 (0.32–0.77)
0.002
AAAGT
32 (3.97)
64 (2.67)
1.19 (0.73–1.92)
0.492
1.19 (0.73–1.93)
0.488
aThe haplotypes order were rs1064034, rs298982, rs62328061, rs9884978, and rs4834698
bObtained in logistic regression models with adjustment for age and gender

False-positive report probability (FPRP) analysis

The obtained significant findings above were further assessed using false-positive report probability (FPRP) analysis (Table 4). At the prior probability of 0.1 and FPRP threshold value of 0.2, the associations between rs1064034 and Wilms tumor risk remained noteworthy in models TA/AA vs. TT and subgroup of children > 18 months in TA/AA vs. TT. Noteworthy results were also found for the GA vs. GG, GA/AA vs. GG, and subgroup of children > 18 months in GA/AA vs. GG. In addition, a significant decrease of Wilms tumor risk was detected in the carrier of 5 vs. 0–4 protective genotypes and subgroup of children > 18 months in 5 vs. 0–4 protective genotypes. Significant findings remained noteworthy in the haplotype TGGGC when compared to reference haplotype TGAAC.
Table 4
False-positive report probability analysis for significant findings
Genotype
OR (95% CI)
Pa
Statistical power b
Prior probability
0.25
0.1
0.01
0.001
0.0001
rs1064034 T > A
 TA vs. TT
0.78 (0.61–0.99)
0.0372
0.899
0.110
0.271
0.804
0.976
0.998
 TA/AA vs. TT
0.77 (0.61–0.97)
0.0237
0.886
0.074
0.194
0.726
0.964
0.996
   > 18
0.66 (0.49–0.87)
0.0033
0.441
0.022
0.063
0.426
0.882
0.987
  Females
0.68 (0.49–0.96)
0.0257
0.544
0.124
0.298
0.824
0.979
0.998
  Stage IV
0.54 (0.29–0.997)
0.049
0.255
0.366
0.634
0.950
0.995
0.999
rs298982 G > A
 GA vs. GG
0.62 (0.46–0.83)
0.0013
0.307
0.013
0.037
0.295
0.809
0.977
 GA/AA vs. GG
0.69 (0.52–0.90)
0.0071
0.571
0.036
0.101
0.552
0.926
0.992
   > 18
0.54 (0.38–0.77)
0.0006
0.134
0.013
0.039
0.308
0.818
0.978
  Female
0.59 (0.38–0.91)
0.0167
0.287
0.149
0.344
0.852
0.983
0.998
  Stage I
0.63 (0.40–0.98)
0.0416
0.399
0.238
0.484
0.912
0.990
0.999
  Stage I + II
0.69 (0.49–0.96)
0.028
0.566
0.129
0.308
0.830
0.980
0.998
  Stage III + IV
0.63 (0.40–0.98)
0.0416
0.400
0.238
0.484
0.911
0.990
0.999
Protective genotypes
 5 vs. 0–4
0.68 (0.52–0.90)
0.0063
0.552
0.033
0.093
0.531
0.919
0.991
   > 18
0.54 (0.38–0.77)
0.0006
0.134
0.013
0.039
0.308
0.818
0.978
  Female
0.60 (0.39–0.93)
0.0216
0.318
0.169
0.379
0.871
0.985
0.999
  Stage I
0.64 (0.41–0.99)
0.0455
0.413
0.248
0.498
0.916
0.991
0.999
  Stage I + II
0.69 (0.50–0.97)
0.0318
0.585
0.140
0.329
0.843
0.982
0.998
  Stage III + IV
0.61 (0.39–0.95)
0.0291
0.338
0.205
0.437
0.895
0.989
0.999
Haplotypes
 TGGGC vs. TGAAC
11.85 (1.37–102.72)
0.025
0.035
0.683
0.866
0.986
0.999
1.000
 AGAGT vs. TGAAC
0.39 (0.19–0.82)
0.012
0.089
0.295
0.557
0.932
0.993
0.999
 TGGGC vs. TGAAC
0.37 (0.18–0.77)
0.008
0.070
0.256
0.508
0.919
0.991
0.999
 TGGGC vs. TGAAC
0.50 (0.32–0.77)
0.002
0.148
0.035
0.099
0.547
0.924
0.992
OR Odds ratio, CI Confidence interval
aChi-square test was used to calculate the genotype frequency distributions
bStatistical power was calculated using the number of observations in each subgroup and the corresponding ORs and P values in this table

Effect of SNPs on gene expression (eQTLs) and splicing (sQTLs)

We further used GTEx to analyze the expression quantitative trait loci (eQTLs) and splicing quantitative trait loci (sQTLs) of rs1064034 and rs298982. Interestingly, rs1064034 was significantly associated with mRNA expression of RP11-384 K6.6 in the whole blood (Fig. 1A) and cells-cultured fibroblasts (Fig. 1B), as well as SNHG8 in cells-cultured fibroblasts (Fig. 1C). We found that the rs1064034 could affect the splicing events of RP11-384 K6.6 (Fig. 1D) and SNHG8 (Fig. 1E) genes in cells-cultured fibroblasts. Similarly, rs298982 was significantly associated with mRNA expression of RP11-384 K6.6 in the whole blood (Fig. 2A) and cells-cultured fibroblasts (Fig. 2B), as well as SNHG8 in cells-cultured fibroblasts (Fig. 2C). SNP rs298982 could also affect the splicing events of RP11-384 K6.6 (Fig. 2D) and SNHG8 (Fig. 2E) genes in cells-cultured fibroblasts.

Discussion

This is the first genetic epidemiological study on the association of genetic variants in the METTL14 gene and Wilms tumor risk. We found that common variants in the METTL14 gene were significantly associated with susceptibility to this malignancy. This study may contribute to uncovering the underlying biology and genetics of Wilms tumor.
METTL14 is a key component of the m6A methyltransferase complex. METTL14 has different roles in different tumors and can be either a cancer promoter or suppressor. Chen et al. [36] identified METTL14 as a tumor suppressor in colorectal cancer. The low METTL14 was significantly associated with poor overall survival. Further functional experiments demonstrated that METTL14 inhibited the progression of colorectal cancer by regulating the production process of m6A-dependent precursor miR-375. Ma et al. [37] found that METTL14 was remarkedly downregulated in hepatocellular carcinoma. The reduced METTL14 expression was significantly associated with unfavorable recurrence-free survival and overall survival. The inhibitory role of METTL14 on hepatocellular carcinoma may be partly attributed to its facilitation of the primary miR-126 maturation in a m6A-dependent manner. METTL14 exerted an oncogenic role in acute myeloid leukemia via mRNA m6A modification [38]. Lang et al. [39] observed that METTL14 was an important driver in EBV-induced oncogenesis. They found that knockdown of METTL14 caused a decreased tumorigenic activity of EBV-transformed cells in the xenograft animal model systems. METTL14 could promote the growth and metastasis of pancreatic cancer by up regulating the m6A level of PERP mRNA [40].
Since the function and mechanism of m6A modification in mammals have not been studied for a long time, the effect of SNPs of m6A modification genes on genetic susceptibility to tumors has been hardly understood. Through adopting a two-stage case-control study, Meng et al. [41] conducted the first study to explore whether m6A gene SNPs could predispose to colorectal cancer in the Chinese population. All the five METTL14 gene SNPs (rs115267066, rs167246, rs2029399, rs298981, and rs441216) failed to show impacts on colorectal cancer risk. By enrolling 898 patients with neuroblastoma and 1734 controls, our group found that the METTL14 gene rs298982 G > A and rs62328061 A > G could significantly reduce the risk of neuroblastoma in children, while rs9884978 G > A and rs4834698 T > C could significantly increase the risk of neuroblastoma [28]. Regarding Wilms tumor, no studies investigating the role of METTL14 gene SNPs were available by far.
In the current study, rs1064034 and rs298982 variant alleles were found to protect from developing Wilms tumor. The combination of five protective genotypes led to a 0.69-fold decrease in the risk of developing Wilms tumor in comparison to 0–4 protective genotypes, indicating the stronger effect of the combined SNPs. It is believed that association studies based on haplotypes of multiple SNPs instead of individual SNP remarkedly strengthen the power for mapping and characterizing disease-causing genes [42, 43]. Thus, we examined whether haplotypes of METTL14 gene are associated with Wilms tumor risk. Expectedly, METTL14 gene haplotypes showed a significantly increased protection against Wilms tumor, indicating the synergistic effects of these SNPs. Genetic variation can modulate gene expression, thereby affecting phenotypes and susceptibility to complex diseases such as Wilms tumor. Here we harnessed the GTEx database to evaluate the effect of SNPs rs1064034 and rs298982 on expression and alternative splicing events of genes. We found that rs1064034 and rs298982 were significantly correlated with the expression and splicing of its nearby genes SNHG8 and RP11-384 K6.6. LncRNA SNHG8 acts as a vital role in tumorigenesis [4448]. Thus, it is biologically possible that changes of the expression and splicing of SNHG8 and RP11-384 K6.6 caused by SNP rs1064034 and rs298982 may influence Wilms tumor risk (Fig. 3). Our results bring new insights into genetic mechanisms of how METTL14 affects Wilms tumor risk. Our findings identify METTL14 gene SNPs as risk markers in pediatric Wilms tumor. These findings not only show the relationship between some METTL14 gene SNPs and Wilms tumor risk but also can help to improve risk stratification strategies for Wilms tumor patients. In all, in-depth mechanism of how METTL14 SNPs affects Wilms tumor risk by regulating the gene expression and splicing pattern awaits to be elucidated. Potential limitations of our study include relatively small sample size, a lack of independent validation, and failure to incorporate other confounders. We also acknowledged that the conclusion obtained here was limited to Chinese. Cautions should be taken when interpreting this conclusion in other populations.

Conclusion

In summary, we demonstrated the significant effects of METTL14 gene SNPs on the risk of Wilms tumor. However, further validation studies with larger sample size and involving different populations are required to strengthen this association.

Acknowledgements

Not applicable.

Declarations

Written informed consent was signed by all subjects’ guardians. The study was carried out based on the principles of the Declaration of Helsinki. Approval of the study protocol was obtained from the institutional review board of Guangzhou Women and Children’s Medical Center (Ethics Approval No: 202016600).
Not applicable.

Competing interests

The author(s) declare that they have no conflict of interest.
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/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Aldrink JH, Heaton TE, Dasgupta R, Lautz TB, Malek MM, Abdessalam SF, et al. Update on Wilms tumor. J Pediatr Surg. 2019;54:390–7.PubMedCrossRef Aldrink JH, Heaton TE, Dasgupta R, Lautz TB, Malek MM, Abdessalam SF, et al. Update on Wilms tumor. J Pediatr Surg. 2019;54:390–7.PubMedCrossRef
2.
Zurück zum Zitat Phelps HM, Kaviany S, Borinstein SC, Lovvorn HN 3rd. Biological Drivers of Wilms Tumor Prognosis and Treatment. Children (Basel). 2018;5:145. Phelps HM, Kaviany S, Borinstein SC, Lovvorn HN 3rd. Biological Drivers of Wilms Tumor Prognosis and Treatment. Children (Basel). 2018;5:145.
3.
Zurück zum Zitat Breslow N, Olshan A, Beckwith JB, Green DM. Epidemiology of Wilms tumor. Med Pediatr Oncol. 1993;21:172–81.PubMedCrossRef Breslow N, Olshan A, Beckwith JB, Green DM. Epidemiology of Wilms tumor. Med Pediatr Oncol. 1993;21:172–81.PubMedCrossRef
4.
Zurück zum Zitat Bao PP, Li K, Wu CX, Huang ZZ, Wang CF, Xiang YM, et al. Recent incidences and trends of childhood malignant solid tumors in Shanghai, 2002-2010. Zhonghua Er Ke Za Zhi. 2013;51:288–94.PubMed Bao PP, Li K, Wu CX, Huang ZZ, Wang CF, Xiang YM, et al. Recent incidences and trends of childhood malignant solid tumors in Shanghai, 2002-2010. Zhonghua Er Ke Za Zhi. 2013;51:288–94.PubMed
6.
Zurück zum Zitat Dome JS, Graf N, Geller JI, Fernandez CV, Mullen EA, Spreafico F, et al. Advances in Wilms tumor treatment and biology: Progress through international collaboration. J Clin Oncol. 2015;33:2999–3007.PubMedPubMedCentralCrossRef Dome JS, Graf N, Geller JI, Fernandez CV, Mullen EA, Spreafico F, et al. Advances in Wilms tumor treatment and biology: Progress through international collaboration. J Clin Oncol. 2015;33:2999–3007.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Spiegl HR, Murphy AJ, Yanishevski D, Brennan RC, Li C, Lu Z, et al. Complications following nephron-sparing surgery for Wilms tumor. J Pediatr Surg. 2020;55:126–9.PubMedCrossRef Spiegl HR, Murphy AJ, Yanishevski D, Brennan RC, Li C, Lu Z, et al. Complications following nephron-sparing surgery for Wilms tumor. J Pediatr Surg. 2020;55:126–9.PubMedCrossRef
8.
Zurück zum Zitat Saltzman AF, Carrasco A Jr, Amini A, Cost NG. Patterns of care and survival comparison of adult and pediatric Wilms tumor in the United States: a study of the National Cancer Database. Urology. 2020;135:50–6.PubMedCrossRef Saltzman AF, Carrasco A Jr, Amini A, Cost NG. Patterns of care and survival comparison of adult and pediatric Wilms tumor in the United States: a study of the National Cancer Database. Urology. 2020;135:50–6.PubMedCrossRef
9.
Zurück zum Zitat Sonn G, Shortliffe LM. Management of Wilms tumor: current standard of care. Nat Clin Pract Urol. 2008;5:551–60.PubMedCrossRef Sonn G, Shortliffe LM. Management of Wilms tumor: current standard of care. Nat Clin Pract Urol. 2008;5:551–60.PubMedCrossRef
10.
Zurück zum Zitat Wong KF, Reulen RC, Winter DL, Guha J, Fidler MM, Kelly J, et al. Risk of adverse health and social outcomes up to 50 years after Wilms tumor: the British childhood Cancer survivor study. J Clin Oncol. 2016;34:1772–9.PubMedCrossRef Wong KF, Reulen RC, Winter DL, Guha J, Fidler MM, Kelly J, et al. Risk of adverse health and social outcomes up to 50 years after Wilms tumor: the British childhood Cancer survivor study. J Clin Oncol. 2016;34:1772–9.PubMedCrossRef
11.
Zurück zum Zitat Haber DA, Buckler AJ, Glaser T, Call KM, Pelletier J, Sohn RL, et al. An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms’ tumor. Cell. 1990;61:1257–69.PubMedCrossRef Haber DA, Buckler AJ, Glaser T, Call KM, Pelletier J, Sohn RL, et al. An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms’ tumor. Cell. 1990;61:1257–69.PubMedCrossRef
12.
Zurück zum Zitat Pelletier J, Bruening W, Li FP, Haber DA, Glaser T, Housman DE. WT1 mutations contribute to abnormal genital system development and hereditary Wilms’ tumour. Nature. 1991;353:431–4.PubMedCrossRef Pelletier J, Bruening W, Li FP, Haber DA, Glaser T, Housman DE. WT1 mutations contribute to abnormal genital system development and hereditary Wilms’ tumour. Nature. 1991;353:431–4.PubMedCrossRef
13.
Zurück zum Zitat Treger TD, Chowdhury T, Pritchard-Jones K, Behjati S. The genetic changes of Wilms tumour. Nat Rev Nephrol. 2019;15:240–51.PubMedCrossRef Treger TD, Chowdhury T, Pritchard-Jones K, Behjati S. The genetic changes of Wilms tumour. Nat Rev Nephrol. 2019;15:240–51.PubMedCrossRef
14.
Zurück zum Zitat Ruteshouser EC, Robinson SM, Huff V. Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes Chromosomes Cancer. 2008;47:461–70.PubMedPubMedCentralCrossRef Ruteshouser EC, Robinson SM, Huff V. Wilms tumor genetics: mutations in WT1, WTX, and CTNNB1 account for only about one-third of tumors. Genes Chromosomes Cancer. 2008;47:461–70.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Turnbull C, Perdeaux ER, Pernet D, Naranjo A, Renwick A, Seal S, et al. A genome-wide association study identifies susceptibility loci for Wilms tumor. Nat Genet. 2012;44:681–4.PubMedPubMedCentralCrossRef Turnbull C, Perdeaux ER, Pernet D, Naranjo A, Renwick A, Seal S, et al. A genome-wide association study identifies susceptibility loci for Wilms tumor. Nat Genet. 2012;44:681–4.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Fu W, Zhuo Z, Hua RX, Fu K, Jia W, Zhu J, et al. Association of KRAS and NRAS gene polymorphisms with Wilms tumor risk: a four-center case-control study. Aging (Albany NY). 2019;11:1551–63.CrossRef Fu W, Zhuo Z, Hua RX, Fu K, Jia W, Zhu J, et al. Association of KRAS and NRAS gene polymorphisms with Wilms tumor risk: a four-center case-control study. Aging (Albany NY). 2019;11:1551–63.CrossRef
17.
Zurück zum Zitat Liu GC, Zhuo ZJ, Zhu SB, Zhu J, Jia W, Zhao Z, et al. Associations between LMO1 gene polymorphisms and Wilms’ tumor susceptibility. Oncotarget. 2017;8:50665–72.PubMedPubMedCentralCrossRef Liu GC, Zhuo ZJ, Zhu SB, Zhu J, Jia W, Zhao Z, et al. Associations between LMO1 gene polymorphisms and Wilms’ tumor susceptibility. Oncotarget. 2017;8:50665–72.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Liu P, Zhuo Z, Li W, Cheng J, Zhou H, He J, et al. TP53 rs1042522 C>G polymorphism and Wilms tumor susceptibility in Chinese children: a four-center case-control study. Biosci Rep. 2019;39:BSR20181891.PubMedPubMedCentralCrossRef Liu P, Zhuo Z, Li W, Cheng J, Zhou H, He J, et al. TP53 rs1042522 C>G polymorphism and Wilms tumor susceptibility in Chinese children: a four-center case-control study. Biosci Rep. 2019;39:BSR20181891.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Ferrara M, Capozzi L, Russo R. Impact of the MTHFR C677T polymorphism on risk of Wilms tumor: case-control study. J Pediatr Hematol Oncol. 2009;31:256–8.PubMedCrossRef Ferrara M, Capozzi L, Russo R. Impact of the MTHFR C677T polymorphism on risk of Wilms tumor: case-control study. J Pediatr Hematol Oncol. 2009;31:256–8.PubMedCrossRef
21.
Zurück zum Zitat Liang Y, Zhan G, Chang KJ, Yang YP, Wang L, Lin J, et al. The roles of m6A RNA modifiers in human cancer. J Chin Med Assoc. 2020;83:221–6.PubMedCrossRef Liang Y, Zhan G, Chang KJ, Yang YP, Wang L, Lin J, et al. The roles of m6A RNA modifiers in human cancer. J Chin Med Assoc. 2020;83:221–6.PubMedCrossRef
23.
Zurück zum Zitat Tao L, Mu X, Chen H, Jin D, Zhang R, Zhao Y, et al. FTO modifies the m6A level of MALAT and promotes bladder cancer progression. Clin Transl Med. 2021;11:e310.PubMedPubMedCentralCrossRef Tao L, Mu X, Chen H, Jin D, Zhang R, Zhao Y, et al. FTO modifies the m6A level of MALAT and promotes bladder cancer progression. Clin Transl Med. 2021;11:e310.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Zhou C, Zhang Z, Zhu X, Qian G, Zhou Y, Sun Y, et al. N6-Methyladenosine modification of the TRIM7 positively regulates tumorigenesis and chemoresistance in osteosarcoma through ubiquitination of BRMS1. EBioMedicine. 2020;59:102955.PubMedPubMedCentralCrossRef Zhou C, Zhang Z, Zhu X, Qian G, Zhou Y, Sun Y, et al. N6-Methyladenosine modification of the TRIM7 positively regulates tumorigenesis and chemoresistance in osteosarcoma through ubiquitination of BRMS1. EBioMedicine. 2020;59:102955.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Huang J, Chen Z, Chen X, Chen J, Cheng Z, Wang Z. The role of RNA N (6)-methyladenosine methyltransferase in cancers. Mol Ther Nucleic Acids. 2021;23:887–96.PubMedPubMedCentralCrossRef Huang J, Chen Z, Chen X, Chen J, Cheng Z, Wang Z. The role of RNA N (6)-methyladenosine methyltransferase in cancers. Mol Ther Nucleic Acids. 2021;23:887–96.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Ma L, Hua RX, Lin H, Zhu J, Fu W, Lin A, et al. The contribution of WTAP gene variants to Wilms tumor susceptibility. Gene. 2020;754:144839.PubMedCrossRef Ma L, Hua RX, Lin H, Zhu J, Fu W, Lin A, et al. The contribution of WTAP gene variants to Wilms tumor susceptibility. Gene. 2020;754:144839.PubMedCrossRef
27.
Zurück zum Zitat Hua RX, Liu J, Fu W, Zhu J, Zhang J, Cheng J, et al. ALKBH5 gene polymorphisms and Wilms tumor risk in Chinese children: a five-center case-control study. J Clin Lab Anal. 2020;34:e23251.PubMedPubMedCentralCrossRef Hua RX, Liu J, Fu W, Zhu J, Zhang J, Cheng J, et al. ALKBH5 gene polymorphisms and Wilms tumor risk in Chinese children: a five-center case-control study. J Clin Lab Anal. 2020;34:e23251.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Zhuo Z, Lu H, Zhu J, Hua RX, Li Y, Yang Z, et al. METTL14 gene polymorphisms confer neuroblastoma susceptibility: an eight-center case-control study. Mol Ther Nucleic Acids. 2020;22:17–26.PubMedPubMedCentralCrossRef Zhuo Z, Lu H, Zhu J, Hua RX, Li Y, Yang Z, et al. METTL14 gene polymorphisms confer neuroblastoma susceptibility: an eight-center case-control study. Mol Ther Nucleic Acids. 2020;22:17–26.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Zhuo ZJ, Liu W, Zhang J, Zhu J, Zhang R, Tang J, et al. Functional polymorphisms at ERCC1/XPF genes confer neuroblastoma risk in Chinese children. EBioMedicine. 2018;30:113–9.PubMedPubMedCentralCrossRef Zhuo ZJ, Liu W, Zhang J, Zhu J, Zhang R, Tang J, et al. Functional polymorphisms at ERCC1/XPF genes confer neuroblastoma risk in Chinese children. EBioMedicine. 2018;30:113–9.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Zhuo Z, Zhou C, Fang Y, Zhu J, Lu H, Zhou H, et al. Correlation between the genetic variants of base excision repair (BER) pathway genes and neuroblastoma susceptibility in eastern Chinese children. Cancer Commun (Lond). 2020;40:641–6.CrossRef Zhuo Z, Zhou C, Fang Y, Zhu J, Lu H, Zhou H, et al. Correlation between the genetic variants of base excision repair (BER) pathway genes and neuroblastoma susceptibility in eastern Chinese children. Cancer Commun (Lond). 2020;40:641–6.CrossRef
31.
Zurück zum Zitat Lin DY, Zeng D, Millikan R. Maximum likelihood estimation of haplotype effects and haplotype-environment interactions in association studies. Genet Epidemiol. 2005;29:299–312.PubMedCrossRef Lin DY, Zeng D, Millikan R. Maximum likelihood estimation of haplotype effects and haplotype-environment interactions in association studies. Genet Epidemiol. 2005;29:299–312.PubMedCrossRef
32.
Zurück zum Zitat Hua RX, Zhuo Z, Ge L, Zhu J, Yuan L, Chen C, et al. LIN28A gene polymorphisms modify neuroblastoma susceptibility: a four-Centre case-control study. J Cell Mol Med. 2020;24:1059–66.PubMedCrossRef Hua RX, Zhuo Z, Ge L, Zhu J, Yuan L, Chen C, et al. LIN28A gene polymorphisms modify neuroblastoma susceptibility: a four-Centre case-control study. J Cell Mol Med. 2020;24:1059–66.PubMedCrossRef
33.
Zurück zum Zitat He J, Wang MY, Qiu LX, Zhu ML, Shi TY, Zhou XY, et al. Genetic variations of mTORC1 genes and risk of gastric cancer in an eastern Chinese population. Mol Carcinog. 2013;52(Suppl 1):E70–9.PubMedCrossRef He J, Wang MY, Qiu LX, Zhu ML, Shi TY, Zhou XY, et al. Genetic variations of mTORC1 genes and risk of gastric cancer in an eastern Chinese population. Mol Carcinog. 2013;52(Suppl 1):E70–9.PubMedCrossRef
34.
Zurück zum Zitat Wacholder S, Chanock S, Garcia-Closas M, El Ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96:434–42.PubMedPubMedCentralCrossRef Wacholder S, Chanock S, Garcia-Closas M, El Ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96:434–42.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Chen X, Xu M, Xu X, Zeng K, Liu X, Sun L, et al. METTL14 suppresses CRC progression via regulating N6-Methyladenosine-dependent primary miR-375 processing. Mol Ther. 2019;28:599–612.PubMedPubMedCentralCrossRef Chen X, Xu M, Xu X, Zeng K, Liu X, Sun L, et al. METTL14 suppresses CRC progression via regulating N6-Methyladenosine-dependent primary miR-375 processing. Mol Ther. 2019;28:599–612.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N (6) -methyladenosine-dependent primary MicroRNA processing. Hepatology. 2017;65:529–43.PubMedCrossRef Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N (6) -methyladenosine-dependent primary MicroRNA processing. Hepatology. 2017;65:529–43.PubMedCrossRef
38.
Zurück zum Zitat Weng H, Huang H, Wu H, Qin X, Zhao BS, Dong L, et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes Leukemogenesis via mRNA m (6) a modification. Cell Stem Cell. 2018;22:191–205 e9.PubMedCrossRef Weng H, Huang H, Wu H, Qin X, Zhao BS, Dong L, et al. METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes Leukemogenesis via mRNA m (6) a modification. Cell Stem Cell. 2018;22:191–205 e9.PubMedCrossRef
39.
Zurück zum Zitat Lang F, Singh RK, Pei Y, Zhang S, Sun K, Robertson ES. EBV epitranscriptome reprogramming by METTL14 is critical for viral-associated tumorigenesis. PLoS Pathog. 2019;15:e1007796.PubMedPubMedCentralCrossRef Lang F, Singh RK, Pei Y, Zhang S, Sun K, Robertson ES. EBV epitranscriptome reprogramming by METTL14 is critical for viral-associated tumorigenesis. PLoS Pathog. 2019;15:e1007796.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wang M, Liu J, Zhao Y, He R, Xu X, Guo X, et al. Upregulation of METTL14 mediates the elevation of PERP mRNA N (6) adenosine methylation promoting the growth and metastasis of pancreatic cancer. Mol Cancer. 2020;19:130.PubMedPubMedCentralCrossRef Wang M, Liu J, Zhao Y, He R, Xu X, Guo X, et al. Upregulation of METTL14 mediates the elevation of PERP mRNA N (6) adenosine methylation promoting the growth and metastasis of pancreatic cancer. Mol Cancer. 2020;19:130.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Meng Y, Li S, Gu D, Xu K, Du M, Zhu L, et al. Genetic variants in m6A modification genes are associated with colorectal cancer risk. Carcinogenesis. 2019;41:8–17. Meng Y, Li S, Gu D, Xu K, Du M, Zhu L, et al. Genetic variants in m6A modification genes are associated with colorectal cancer risk. Carcinogenesis. 2019;41:8–17.
42.
Zurück zum Zitat Akey J, Jin L, Xiong M. Haplotypes vs single marker linkage disequilibrium tests: what do we gain? Eur J Hum Genet. 2001;9:291–300.PubMedCrossRef Akey J, Jin L, Xiong M. Haplotypes vs single marker linkage disequilibrium tests: what do we gain? Eur J Hum Genet. 2001;9:291–300.PubMedCrossRef
43.
44.
Zurück zum Zitat Dong J, Teng F, Guo W, Yang J, Ding G, Fu Z. lncRNA SNHG8 promotes the tumorigenesis and metastasis by sponging miR-149-5p and predicts tumor recurrence in hepatocellular carcinoma. Cell Physiol Biochem. 2018;51:2262–74.PubMedCrossRef Dong J, Teng F, Guo W, Yang J, Ding G, Fu Z. lncRNA SNHG8 promotes the tumorigenesis and metastasis by sponging miR-149-5p and predicts tumor recurrence in hepatocellular carcinoma. Cell Physiol Biochem. 2018;51:2262–74.PubMedCrossRef
45.
Zurück zum Zitat Qu X, Li Y, Wang L, Yuan N, Ma M, Chen Y. LncRNA SNHG8 accelerates proliferation and inhibits apoptosis in HPV-induced cervical cancer through recruiting EZH2 to epigenetically silence RECK expression. J Cell Biochem. 2020;121:4120–9.PubMedCrossRef Qu X, Li Y, Wang L, Yuan N, Ma M, Chen Y. LncRNA SNHG8 accelerates proliferation and inhibits apoptosis in HPV-induced cervical cancer through recruiting EZH2 to epigenetically silence RECK expression. J Cell Biochem. 2020;121:4120–9.PubMedCrossRef
46.
Zurück zum Zitat Song H, Song J, Lu L, Li S. SNHG8 is upregulated in esophageal squamous cell carcinoma and directly sponges microRNA-411 to increase oncogenicity by upregulating KPNA2. Onco Targets Ther. 2019;12:6991–7004.PubMedPubMedCentralCrossRef Song H, Song J, Lu L, Li S. SNHG8 is upregulated in esophageal squamous cell carcinoma and directly sponges microRNA-411 to increase oncogenicity by upregulating KPNA2. Onco Targets Ther. 2019;12:6991–7004.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Song Y, Zou L, Li J, Shen ZP, Cai YL, Wu XD. LncRNA SNHG8 promotes the development and chemo-resistance of pancreatic adenocarcinoma. Eur Rev Med Pharmacol Sci. 2018;22:8161–8.PubMed Song Y, Zou L, Li J, Shen ZP, Cai YL, Wu XD. LncRNA SNHG8 promotes the development and chemo-resistance of pancreatic adenocarcinoma. Eur Rev Med Pharmacol Sci. 2018;22:8161–8.PubMed
48.
Zurück zum Zitat Zhen Y, Ye Y, Wang H, Xia Z, Wang B, Yi W, et al. Knockdown of SNHG8 repressed the growth, migration, and invasion of colorectal cancer cells by directly sponging with miR-663. Biomed Pharmacother. 2019;116:109000.PubMedCrossRef Zhen Y, Ye Y, Wang H, Xia Z, Wang B, Yi W, et al. Knockdown of SNHG8 repressed the growth, migration, and invasion of colorectal cancer cells by directly sponging with miR-663. Biomed Pharmacother. 2019;116:109000.PubMedCrossRef
Metadaten
Titel
METTL14 gene polymorphisms decrease Wilms tumor susceptibility in Chinese children
verfasst von
Zhenjian Zhuo
Rui-Xi Hua
Huizhu Zhang
Huiran Lin
Wen Fu
Jinhong Zhu
Jiwen Cheng
Jiao Zhang
Suhong Li
Haixia Zhou
Huimin Xia
Guochang Liu
Wei Jia
Jing He
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2021
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-021-09019-5

Weitere Artikel der Ausgabe 1/2021

BMC Cancer 1/2021 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

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