Skip to main content

METTL14 gene polymorphisms decrease Wilms tumor susceptibility in Chinese children

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.

Peer Review reports

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 [7,8,9]. 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 [11,12,13,14,15]. Apart from these, genetic association analyses in case-control studies also unveiled some Wilms tumor susceptibility loci [16,17,18,19]. 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 [23,24,25]. 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 [28,29,30]. 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

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

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

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

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.

Fig. 1
figure 1

Functional relevance of rs1064034 on gene expression and splicing events in GTEx database. rs1064034 was significantly associated with RP11-384 K6.6 level in the A whole blood (P = 9.9*10−14) and B cells-cultured fibroblasts (P = 3.5*10−12) as well as CSNHG8 mRNA level in the cells-cultured fibroblasts (P = 1.8*10−5). rs1064034 can affect the splicing events of DRP11-384 K6.6 (P = 2.3*10−7) and ESNHG8 (P = 4.1*10−5) genes in cells-cultured fibroblasts

Fig. 2
figure 2

Functional relevance of rs298982 on gene expression and splicing events in GTEx database. rs298982 was significantly associated with RP11-384 K6.6 level in the A whole blood (P = 3.9*10−9) and B cells-cultured fibroblasts (P = 9.4*10−9) as well as CSNHG8 mRNA level in the cells-cultured fibroblasts (P = 1.8*10−6). rs1064034 can affect the splicing events of DRP11-384 K6.6 (P = 8.7*10− 7) and ESNHG8 (P = 4.3*10− 6) 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 [44,45,46,47,48]. 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.

Fig. 3
figure 3

Possible mechanism of how SNPs rs1064034 and rs298982 confer to Wilms tumor risk

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.

Availability of data and materials

All data and material are available from the corresponding author on reasonable request. The datasets generated or analyzed during the current study are not publicly available but are available with the corresponding author and can be provided on reasonable request.

Abbreviations

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

References

  1. 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.

    Article  PubMed  Google Scholar 

  2. Phelps HM, Kaviany S, Borinstein SC, Lovvorn HN 3rd. Biological Drivers of Wilms Tumor Prognosis and Treatment. Children (Basel). 2018;5:145.

    Google Scholar 

  3. Breslow N, Olshan A, Beckwith JB, Green DM. Epidemiology of Wilms tumor. Med Pediatr Oncol. 1993;21:172–81.

    Article  CAS  PubMed  Google Scholar 

  4. 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  Google Scholar 

  5. Hohenstein P, Pritchard-Jones K, Charlton J. The yin and yang of kidney development and Wilms’ tumors. Genes Dev. 2015;29:467–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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.

    Article  PubMed  Google Scholar 

  8. 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.

    Article  PubMed  Google Scholar 

  9. Sonn G, Shortliffe LM. Management of Wilms tumor: current standard of care. Nat Clin Pract Urol. 2008;5:551–60.

    Article  PubMed  Google Scholar 

  10. 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.

    Article  CAS  PubMed  Google Scholar 

  11. 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.

    Article  CAS  PubMed  Google Scholar 

  12. 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.

    Article  CAS  PubMed  Google Scholar 

  13. Treger TD, Chowdhury T, Pritchard-Jones K, Behjati S. The genetic changes of Wilms tumour. Nat Rev Nephrol. 2019;15:240–51.

    Article  PubMed  Google Scholar 

  14. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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.

    Article  CAS  Google Scholar 

  17. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  18. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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.

    Article  CAS  PubMed  Google Scholar 

  20. Cao G, Li HB, Yin Z, Flavell RA. Recent advances in dynamic m6A RNA modification. Open Biol. 2016;6:160003.

    Article  PubMed  PubMed Central  Google Scholar 

  21. 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.

    Article  CAS  PubMed  Google Scholar 

  22. Meyer KD, Jaffrey SR. Rethinking m (6) a readers, writers, and erasers. Annu Rev Cell Dev Biol. 2017;33:319–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  25. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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.

    Article  CAS  PubMed  Google Scholar 

  27. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  30. 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.

    Article  Google Scholar 

  31. 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.

    Article  CAS  PubMed  Google Scholar 

  32. 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.

    Article  CAS  PubMed  Google Scholar 

  33. 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.

    Article  CAS  PubMed  Google Scholar 

  34. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Carithers LJ, Moore HM. The genotype-tissue expression (GTEx) project. Biopreserv Biobank. 2015;13:307–8.

    Article  PubMed  PubMed Central  Google Scholar 

  36. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  37. 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.

    Article  CAS  PubMed  Google Scholar 

  38. 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.

    Article  CAS  PubMed  Google Scholar 

  39. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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.

    Google Scholar 

  42. 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.

    Article  CAS  PubMed  Google Scholar 

  43. Manolio TA, Brooks LD, Collins FS. A HapMap harvest of insights into the genetics of common disease. J Clin Invest. 2008;118:1590–605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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.

    Article  CAS  PubMed  Google Scholar 

  45. 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.

    Article  CAS  PubMed  Google Scholar 

  46. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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.

    CAS  PubMed  Google Scholar 

  48. 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.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No: 82003523, 81803320), Natural Science Foundation of Guangdong Province (No: 2021A1515010860), and Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease (No: 2019B030301004).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: HX, GL and JH; Data curation: JH; Formal analysis: ZZ and JH; Funding acquisition: RH, WF, HX, and JH; Investigation: ZZ, RH, HZ, WJ, HL, WF and JH; Methodology: JHZ and JH; Project administration: JH; Resources: WJ, WF, JC, JZ, SL, HZ and GL; Software: ZZ and JH; Supervision: WJ and JH; Validation: ZZ and RH; Visualization: ZZ; Roles/Writing - original draft: ZZ, JHZ and JH; Writing - review & editing: All authors. All authors had given final approval of the version to be published.

Corresponding authors

Correspondence to Wei Jia or Jing He.

Ethics declarations

Ethics approval and consent to participate

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).

Consent for publication

Not applicable.

Competing interests

The author(s) declare that they have no conflict of interest.

Additional information

Publisher’s Note

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

Supplementary Information

Additional file 1: Table S1

. Frequency distribution of selected variables in Wilms tumor patients and cancer-free controls.

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhuo, Z., Hua, RX., Zhang, H. et al. METTL14 gene polymorphisms decrease Wilms tumor susceptibility in Chinese children. BMC Cancer 21, 1294 (2021). https://doi.org/10.1186/s12885-021-09019-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-021-09019-5

Keywords