Skip to main content

Six polymorphisms in the lncRNA H19 gene and the risk of cancer: a systematic review and meta-analysis

Abstract

Background

Numerous studies have demonstrated long noncoding RNA (lncRNA) play an important role in the occurrence and progression of cancer, and single nucleotide polymorphisms (SNPs) located in lncRNA are considered to affect cancer suspensibility. Herein, a meta-analysis was carried out to better assess the relationship of H19 polymorphisms and cancer susceptibility.

Methods

A literature search was conducted through using PubMed, EMBASE, and Web of Science databases to obtain relevant publications before Aug 23, 2022. The reference lists of the retrieved studies were also investigated to identify additional relevant articles. The pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated to appraise the risk of various cancers.

Results

There appeared to be a remarkable correlation between the rs2107425 variation and decreased cancer risk among Caucasians. Nevertheless, the rs217727 polymorphism was significantly associated with an increased risk of lung cancer, hepatocellular carcinoma and oral squamous cell carcinoma. Also, we found a significant correlation between the rs2839698 polymorphism and increased cancer risk among Asians, gastric cancer, hepatocellular carcinoma, hospital-based control and larger simple size subgroups, respectively. Similarly, the rs3741219 mutation was notably related to cancer risk in higher quality score. As for rs3024270 polymorphism, the homozygous model was markedly linked to cancer risk in overall analysis and population-based controls. There was no significant association between the rs3741216 polymorphism and cancer risk.

Conclusion

H19 rs2839698 and rs3024270 were closely associated with overall cancer risk. H19 rs2107425 was related to lower cancer risk among Caucasians, while the rs2839698 was related to increased cancer risk among Asians. Our results supported that H19 SNPs were significantly correlated with cancer risk.

Peer Review reports

Introduction

Cancer has been the second biggest cause of mortality worldwide, seriously endangering public health and increasing economic burden on society [1]. In 2023, 1,918,030 new cancer cases and 609,360 cancer mortalities are estimated to occur in the United States. And prostate, lung, and colorectal cancers account for 48% of all male incident cases, while 51% of all female incident cases are diagnosed with breast, lung, and colorectal cancers [2]. Although the specific pathological mechanism of tumorigenesis still remains unclear, cancer is considered as a complex and multifactorial disease that results from the interaction of environmental and genetic risk factors, such as high-calorie diet, smoking, excessive drinking, obesity, hypertension, diabetes [3,4,5]. Recent advances in cancer diagnosis and treatment, including multifunctional nanomaterials combined with imaging probes and drugs, nanomedicine products and therapeutic vaccines are improving options for cancer patients [6,7,8]. On the other hand, preference heterogeneity between patients indicates that tradeoffs between survival benefits and long-term physical, emotional, cognitive, and functional side effects should be carefully considered in treatment decision-making [9]. At present, genome-wide association studies (GWAS) have identified a strong association of several common single nucleotide polymorphisms (SNPs) with cancer risk [10, 11]. Certain genetic SNPs were found to be related to cancer risk, including miR-143/145, CASP9, CASP10 and IL-1β [12,13,14]. In addition, functional SNPs are present in lncRNA genes and influence gene expression and function through various means, and then result in the occurrence and progression of cancer [15].

Being widely transcribed in the human genome, long non-coding RNAs (lncRNAs) are defined as single stranded non-coding RNAs with a length of more than 200 base pairs and no open reading frames, thereby lacking of protein-coding function, although some of them may produce small functional peptides [16]. LncRNAs take part in numerous cellular processes by interacting with cellular molecules, such as DNAs, RNAs, or proteins [17]. At the levels of epigenetic, transcriptional, and post-transcriptional modifications, they can regulate gene expression via different mechanisms, including chromatin remodeling induction, alternative splicing, intranuclear transport, production of miRNA sponges, and transcriptional interference [18,19,20,21,22]. Interestingly, lncRNAs paly crucial regulatory roles in a variety of physiological and pathological processes and cancer biology, including cell proliferation, differentiation, apoptosis, and carcinogenesis progression [23,24,25,26,27]. It has been found that lncRNAs are dysregulated in various types of cancer, which contributes to tumorigenesis and development of tumors by affecting the expression of oncogenes or tumor suppressors. Generally, lncRNAs are thought to have prospective clinical implications and to be appraised as independent novel biomarkers for diagnosis and prognosis in human cancer treatment [28,29,30].

As a critical maternally imprinted gene, lncRNA H19 was initially discovered in the 1990s [31]. The H19 gene, possessing five exons and four introns, encodes a 2.3-kb long, capped, spliced, and polyadenylated noncoding RNA, of which the transcript is highly conserved at a cluster with the insulin-like growth factor 2 (IGF2) locus on human chromosome 11p15.5, and plays an essential role in embryonic development and growth control [32,33,34,35]. It has been reported that the aberrant expression of H19 was implicated in various types of cancer, including breast, lung, esophageal, gastric, pancreatic, colorectal, liver, bladder and cervical cancer. H19 acts as an oncogene or a suppressor gene, which may be attributed to the heterogeneity of different types of cancer [36,37,38]. Previous researches have shown that H19 gene polymorphisms are markedly associated with malignancies, however, the results were controversial and inconsistent. Therefore, the aim of this meta-analysis was to accurately examine the correlation between H19 polymorphisms and cancer susceptibility.

Materials and methods

Literature search strategy

Eligible studies were retrieved from the PubMed, EMBASE, and Web of Science electronic databases up to Aug 23, 2022. Our search strategy included the main terms for: (H19 or long Noncoding RNA H19 or lncRNA H19) and (polymorphism or genotype or SNP) and (carcinoma or neoplasm or cancer or tumor). At the same time, we manually screened out the relevant potential articles in the references extracted.

Selection and exclusion criteria

Inclusion criteria are as follows: (1) case-control studies investigated the relationship between H19 polymorphisms and the risk of cancer; (2) the histopathological diagnosis of cancer patients was clearly defined; the control group did not have any history of cancer; (3) sufficient data on genotype distribution of H19 polymorphisms was applied to calculate the odds ratio (OR) and 95% confidence interval (CI).

The exclusion criteria were as follows: (1) abstract, case reports, comment, editorials and review; (2) duplication of the previous reports; (3) lack of the full text or main genotyping data; (4) non-case-control or cohort design studies.

Data extraction

Two investigators separately conducted literature screening, data extraction, literature quality evaluation, and any disagreements that could be resolved through discussion or a third analyst. The relevant information independently extracted by two investigators included the following information from each study: first author, year of publication, country of the population, ethnicity, source of controls, genotyping methods, cancer types, sample size and P value of (HWE).

The Newcastle-Ottawa scale (NOS) was adopted to assess the process in terms of queue selection, comparability of queues, and evaluation of results [39]. A study with a score of at least six was considered as a high-quality literature. Higher NOS scores showed higher literature quality.

Statistical analysis

All data analysis was conducted using Stata16.0 software (Stata Corp LP, TX, USA). Odds ratio (OR) and 95% confidence intervals (CIs) were used to evaluate the association between lncRNA H19 polymorphisms and various cancers. After that, the heterogeneity test was conducted. When P ≥ 0.05 or I2 < 50% was performed, it indicated that there was no obvious heterogeneity, and the fixed-effect pattern should be applied for a merger. Otherwise, the random-effect model was used. Results were considered significant statistically when the p-value less than 0.05. Subgroup analysis was implemented to determine the source of heterogeneity. Additionally, sensitivity analysis was performed to assess the impact of each individual study on overall results. The Begg’s rank correlation test and Egger’s linear regression test were used to verify the publication bias among these studies. If P < 0.05 indicates obvious publication bias.

False-positive report probability (FPRP) analysis

The probability of meaningful relationships between H19 SNPs and cancer risk can be determined through carrying out the FPRP analysis [40]. In order to investigate the remarkable associations observed in the meta-analysis, we adopted prior probabilities of 0.25, 0.1, 0.01, 0.001, and 0.0001 and computed the FPRP values as described previously. The association that reached the FPRP threshold of < 0.2 was considered significant.

Results

Process of study selection and description of qualified studies

As shown in Fig. 1, the initial 472 studies were retrieved by databases of PubMed (n = 229), Embase (n = 76), Web of science (n = 166). After eliminating 152 duplicate articles, 191 additional publications were excluded by screening the abstract and title. Among these, 147 articles were reviews, letters, conference abstracts, meta-analysis, notes, editorials and short surveys, and 44 articles focused on animal or vitro experiment. After careful review of the full texts, 88 articles were further excluded due to the following reasons: 30 articles were involved with other genes or other SNPs of H19, 45 studies were not relevant to cancer and 13 studies had no available data. Finally, the remaining 40 eligible articles were included in this analysis [41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80].

Fig. 1
figure 1

Flow diagram of the eligible study selection process

Through literature search and selection, a total of 40 eligible articles embodying 95 studies were embodying in our study, which included 13 studies for rs2107425, 30 studies for rs217727, 26 studies for rs2839698, 10 studies for rs3741219, 12 studies for rs3024270, and 4 studies for rs3741216 polymorphisms. One article referred to two independent case-control studies, and thus the study was regarded as two separate estimates [44]. Among the included studies, 30 studies were from China, four studies from Iran, two studies form European countries, two studies from Egypt, two studies from the mixed countries, and one study from America. At the same time, 34 studies were conducted in the Asian descent, five studies were conducted in the Caucasian descent and two studies were conducted in the African descent. Thirteen of the studies focused on population-based controls and 27 on hospital-based controls. If the number of different cancer types is less than 1, the cancer type is classified into other cancer subgroup. The detailed characteristics of selected studies are illustrated in Table 1, such as cancer type, genotyping method, sample size, distributions of genotype frequency and Hardy-Weinberg equilibrium. The NOS score of all articles ranged from 6 to 8, implying that all included studies were of high quality.

Table 1 Characteristics of all studies included in the meta-analysis

Correlation between rs2107425 C/T polymorphism and cancer risk

Thirteen relevant studies with 11,972 cancer patients and 17,128 controls were examined for the association between the rs2107425 polymorphism and cancer risk. Compared with the wild-type CC homozygote, the genotypes of rs2107425 were not linked to cancer risk in overall analyses (T vs. C: OR = 0.98, 95%CI = 0.91–1.06, P = 0.595; TT vs. CC: OR = 1.01, 95%CI = 0.88–1.17, P = 0.846; TC vs. CC: OR = 0.96, 95%CI = 0.85–1.07, P = 0.438). Similarly, no relationships were detected in the dominant and recessive models (TT + TC vs. CC: OR = 0.97, 95%CI = 0.87–1.08, P = 0.543; TT vs. TC + CC: OR = 0.98, 95%CI = 091-1.06, P = 0.651; Table 2; Fig. 2). Stratification analysis by ethnicity showed the rs2107425 variation significantly reduced cancer risk among Caucasians (T vs. C: OR = 0.91, 95% CI = 0.85 − 0.7, P = 0.006; CT vs. CC: OR = 0.83, 95% CI = 0.73–0.94, P = 0.003; OR = 0.85, 95% CI = 0.76–0.94, P = 0.003), which might be a protective factor in the Caucasian population. Also, we found a significant association of rs2107425 variant with cancer risk under the heterozygote models in hospital-based subgroup (CT vs. CC: OR = 1.18, 95% CI = 1.00-1.39, P = 0.049) and population-based source of controls (CT vs. CC: OR = 0.87, 95% CI = 0.78–0.97, P = 0.016, Table 2). There was significant association between the rs2107425 variant and elevated risk of CRC (T vs. C: OR = 3.15, 95%CI = 1.51–6.57, P = 0.002; TT vs. CC: OR = 10.40, 95%CI = 0.1.25–86.4, P = 0.030; TC vs. CC: OR = 2.84, 95%CI = 1.11–7.32, P = 0.030; TT + TC vs. CC: OR = 3.60, 95%CI = 1.46–8.88, P = 0.005). The heterozygote and recessive models of rs2107425 notably decreased the risk of hepatocellular carcinoma (TT vs. CC: OR = 0.61, 95%CI = 0.41–0.90, P = 0.012; TT vs. CC + TC: OR = 0.59, 95%CI = 0.41–0.85, P = 0.004, Table 2). Heterogeneity test results suggested that heterogeneity existed in all five genetic models of overall analyses. Heterogeneity was not observed under the allelic, homozygote, and recessive models in Caucasians subgroup.

Table 2 Summary ORs and 95% CIs of H19 SNPs and risk of cancer
Fig. 2
figure 2

Forest plots for the association between H19 rs2107425 polymorphism and cancer risk in five models. A: allele model; B: dominant model; C: heterozygote model; D: homozygote model; E: recessive model

Correlation between rs217727 G/A polymorphism and cancer risk

Intriguingly, we obtained thirty studies about the relationship between rs217727 polymorphism and cancer risk with 14,215 patients and 20,247 controls. Overall, the rs217727 polymorphism was not significantly correlated with cancer risk (Table 2; Fig. 3). The allelic, homozygote, dominant and recessive models of rs217727 notably increased the risk of lung cancer (A vs. G: OR = 1.16, 95% CI = 1.06–1.27, P = 0.002; AA vs. GG: OR = 1.38, 95% CI = 1.14–1.67, P = 0.001; AA + GA vs. GG: OR = 1.16, 95% CI = 1.01–1.33, P = 0.031; AA vs. GG + GA: OR = 1.31, 95% CI = 1,03-1.66, P = 0.028) and oral squamous cell carcinoma (A vs. G: OR = 1.31, 95% CI = 1.14–1.50, P = 0.000; AA vs. GG: OR = 1,89, 95% CI = 1.18-3.00, P = 0.008; GA vs. GG: OR = 1.27, 95% CI = 1.07–1.50, P = 0.006; AA + GA vs. GG: OR = 1.36, 95% CI = 1.16–1.60, P = 0.000; AA vs. GG + GA: OR = 1.67, 95% CI = 1.04–2.68, P = 0.035, Table 2). Additionally, the rs217727 mutation significantly decreased the risk of hepatocellular carcinoma (GA vs. GG: OR = 0.68, 95% CI = 0.49–0.93, P = 0.017; AA vs. GG + GA: OR = 0.73, 95% CI = 0.54-1.00, P = 0.048, Table 2), suggesting that the rs217727 mutation may be an important protective factor for liver cancer, but a key risk factor for lung cancer and oral squamous cell carcinoma. The pooled results indicated that the homozygote and recessive models of rs217727 have a positive association with cancer risk in larger sample size (AA vs. GG: OR = 1.17, 95% CI = 1.02–1.33, P = 0.022; AA vs. GG + GA: OR = 1.14, 95% CI = 1.03–1.28, P = 0.015, Table 2). Heterogeneity was shown to exist in all five gene models, and results demonstrated that heterogeneity significantly decreased or disappeared in lung cancer and oral squamous cell carcinoma.

Fig. 3
figure 3

Forest plots for the association between H19 rs217727 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

Correlation between rs2839698 G/A polymorphism and cancer risk

A total of twenty-six studies with 12,413 cancer patients and 18,650 controls were included to examine the association between H19 SNP rs2839698 and cancer risk. The rs2839698 polymorphism remarkably enhanced the risk of cancer in the allelic, homozygote, dominant and recessive models (A vs. G: OR = 1.10, 95% CI = 1.01–1.20, P = 0.039; AA vs. GG: OR = 1.29, 95% CI = 1.09–1.52, P = 0.003; AA + GA vs. GG: OR = 1.18, 95% CI = 1.01–1.23, P = 0.036; AA vs. GG + GA: OR = 1.18, 95% CI = 1.01–1.39, P = 0.042, Table 2; Fig. 4). Next, stratification analyses by cancer type showed the rs2839698 mutation significantly increased the risk of gastric cancer (A vs. G: OR = 1.33, 95% CI = 1.13–1.56, P = 0.000; AA vs. GG: OR = 1.76, 95% CI = 1.26–2.46, P = 0.001; AA + GA vs. GG: OR = 1.27, 95% CI = 1.03–1.57, P = 0.024; AA vs. GG + GA: OR = 1.74, 95% CI = 1.27–2.40, P = 0.001), hepatocellular cancer (A vs. G: OR = 1.17, 95% CI = 1.03–1.34, P = 0.015; GA vs. GG: OR = 1.30, 95% CI = 1.08–1.56, P = 0.006; AA + GA vs. GG: OR = 1.29, 95% CI = 1.08–1.93, P = 0.005), renal cell carcinoma and ovarian cancer, leukemia and lymphoma (Table 2). Similarly, a positive association was detected between the allelic, homozygous, and dominant models and cancer susceptibility in the Asian descent (A vs. G: OR = 1.10, 95% CI = 1.00-1.21, P = 0.041; AA vs. GG: OR = 1.30, 95% CI = 1.09–1.54, P = 0.003; AA + GA vs. GG: OR = 1.12, 95% CI = 1.02–1.24, P = 0.024, Table 2). When stratifying by source of control, quality score and sample size, the significantly increased cancer risk was discovered in hospital-based control (AA vs. GG: OR = 1.30, 95% CI = 1.07–1.59, P = 0.009; AA + GA vs. GG: OR = 1.14, 95% CI = 1.02–1.28, P = 0.025), population-based control (AA vs. GG + GA: OR = 1.28, 95% CI = 1.02–1.59, P = 0.032) and large simple size (A vs. G: OR = 1.11, 95% CI = 1.01–1.21, P = 0.030; AA vs. GG: OR = 1.28, 95% CI = 1.07–1.53, P = 0.006; AA vs. GG + GA: OR = 1.25, 95% CI = 1.09–1.45, P = 0.002, Table 2). Heterogeneity results suggested that heterogeneity consisted in the five genetic models of overall analysis. Interestingly, we found that heterogeneity notably diminish or disappear in hepatocellular carcinoma, bladder, gastric, and lung cancer.

Fig. 4
figure 4

Forest plots for the association between H19 rs2839698 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

Correlation between rs3741219 A/G polymorphism and cancer risk

To explore the association between H19 rs3741219 polymorphism and cancer risk, we included 10 studies with 5305 patients and 6974 controls. Compared with AA + GA genotype, the GG allele of rs3741219 polymorphism was correlated with cancer susceptibility in overall analysis (AA vs. GG + GA: OR = 1.14, 95% CI = 1.01–1.29; P = 0.037, Table 2; Fig. 5). Stratified analyses indicated that the rs3741219 mutant remarkably enhanced the risk of hepatocellular carcinoma and ovarian cancer, but also decreased the risk of Glioma tumor. We next performed stratification analysis by source of control and sample size, the pooled results indicated no association between 3,741,219 polymorphism and cancer risk. Beyond that, subgroup analyses by quality score strongly showed an elevated cancer risk in higher quality score (G vs. A: OR = 1.11, 95% CI = 1.02–1.21, P = 0.015; GG vs. AA: OR = 1.26, 95% CI = 1.03–1.53, P = 0.022; GG + GA vs. AA: OR = 1.12, 95% CI = 1.00-1.26, P = 0.042; GG vs. AA + GA: OR = 1.19, 95% CI = 0.98–1.43, P = 0.038, Table 2). It manifested that heterogeneity mainly appeared in the five gene models of overall analysis and Asian population. Moreover, there was no heterogeneity existing in population-based and higher quality score.

Fig. 5
figure 5

Forest plots for the association between H19 rs3741219 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

Correlation between rs3024270 C/G polymorphism and cancer risk

Through integrating 12 potential studies embodying 5402 patients and 9159 controls, we found a significant relationship of rs3024270 polymorphism with cancer risk under homozygous model (GG vs. CC: OR = 1.12, 95% CI = 1.01–1.24, P = 0.025, Table 2; Fig. 6). The homozygous and recessive models of rs3024270 were significantly correlated with the increased risk of colorectal cancer (GG vs. CC: OR = 1.28, 95% CI = 1.01–1.61, P = 0.041; GG vs. CC + GC: OR = 1.29, 95% CI = 1.04–1.58, P = 0.019). There was no significant association between the rs3024270 polymorphism and cancer susceptibility in stratification analysis by ethnicity and quality score. We found that the rs3024270 polymorphism was positively related to cancer risk in hospital-based controls under the homozygote model (GG vs. CC: OR = 1.42, 95% CI = 1.05–1.93; P = 0.025, Table 2). Except for the recessive model (I2 = 65.3%, P = 0.001), there was no heterogeneity in other models.

Fig. 6
figure 6

Forest plots for the association between H19 rs3024270 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

Correlation between rs3741216 A/T polymorphism and cancer risk

In general, four eligible studies with 2049 patients and 1808 controls were included to detect the relation between rs2107425 polymorphism and cancer risk. The pooled results suggested that the rs2107425 polymorphism was not related to cancer risk in five genetic models (T vs. A: OR = 1.66, 95% CI = 0.87–3.18, P = 0.127; TT vs. AA: OR = 1.66, 95% CI = 0.85–1.56, P = 0.348; AT vs. AA: OR = 0.91, 95% CI = 0.78–1.06, P = 0.236; AT + TT vs. AA: OR = 0.95, 95% CI = 0.82–1.10, P = 0.471; TT vs. AA + AT: OR = 2.42, 95% CI = 0.66–8.83, P = 0.181, Table 2; Fig. 7). Similarly, when stratifying analyses by ethnicity, cancer type, quality score, and source of control, we did find any correlation between the rs3741216 polymorphism and cancer risk. The result of heterogeneity test exhibited I2 = 95.9 and 95.7, implying that heterogeneity clearly exists in both homologous and recessive models, and thus random-effects model was used to examine the correlation.

Fig. 7
figure 7

Forest plots for the association between H19 rs3741216 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

FPRP results

We investigated determinants of FPRP across a range of probabilities to determine whether a given association between H19 SNPs and cancer risk is deserving of attention or is noteworthy. In this respect, we found that our main results were further supported by FPRP analysis. As shown in Table 3, with a prior probability < 0.25, the H19 rs2839698 polymorphism was associated with the risk of cancer under allele, homozygote, dominant and recessive models. Similarly, with a prior probability of 0.25, the homozygote model of H19 rs3024270 polymorphism was associated with cancer risk and the recessive model of H19 rs3024270 polymorphis was associated with cancer risk (P < 0.2).

Table 3 False-positive report probability analysis of the noteworthy results

Sensitivity analysis and publication bias

Sensitivity analysis was conducted by eliminating each individual study. As shown in Fig. 8, the pooled OR and 95% CI were not materially changed, indicating that our results were relatively robust. After excluding several studies inconsistent with HWE, we found substantial alteration under the allele and heterozygous models in rs3741216 polymorphism (allelic: I2 = 0.0%, P(heterogeneity) = 0.649; heterozygous: I2 = 0.0, P(heterogeneity) = 0.678; homozygous: I2 = 0.0%, P(heterogeneity) = 0.737; dominant: I2 = 0.0%, P(heterogeneity) = 0.681; recessive: I2 = 0.0%, P(heterogeneity) = 0.708, Table 4). Other three gene polymorphisms were not substantially changed. In addition, funnel plot was symmetrical, and no evident publication bias was observed by using the Begg’s test and Egger’s test (Table 5; Fig. 9).

Table 4 After excluding studies inconsistent with HWE, the associations between four H19 polymorphisms and cancer risk under five genetic models
Table 5 Publication bias of the five genetic models for H19 gene polymorphisms
Fig. 8
figure 8

Sensitivity analysis for H19 rs2839698 polymorphism and cancer risk in five models. A: allele model; B: homozygote model; C: heterozygote model; D: dominant model; E: recessive model

Fig. 9
figure 9

Begg’s funnel plot and Egger’s linear regression plot for detecting the publication bias in rs2839698 polymorphism. (a1) Begg’s funnel plot and (b1) Egger’s linear regression plot in the allele model; (a2) Begg’s funnel plot and (b2) Egger’s linear regression plot in the homozygote model; (a3) Begg’s funnel plot and (b3) Egger’s linear regression plot in the homozygote model; (a4) Begg’s funnel plot and (b4) Egger’s linear regression plot in the heterozygote model; (a5) Begg’s funnel plot and (b5) Egger’s linear regression plot in the recessive model

Discussion

Cancer is one of the leading causes of mortality, seriously affecting public health all over the world. However, the pathogenesis of cancer remains poorly explicit. It is widely accepted that cancer may be influenced by genetic mutations [81]. As a newly identified non-coding RNAs, lncRNAs are extensively present in human genome. Numerous studies have confirmed that lncRNAs play essential roles in diverse biological activities, such as cell cycle processes, epigenetic regulation, transcriptional regulation, stress response and pluripotency maintenance [16, 18]. A large number of SNPs located in the lncRNAs may affect gene expression and function by altering its secondary structure or the targeted microRNAs, ultimately, leading to the occurrence and progression of cancer [82,83,84].

H19 belongs to a class of maternally expressed lncRNA at 2.3 kb length, which is located at imprinted region on chromosome 11p15.5. Differentially methylated region (DMR) usually refers to the upstream of the transcription initiation site of H19, which servers a vital role in the regulation of H19/IGF2 expression [85, 86]. It has been reported that H19 expression is prominently decreased after birth, and only exhibits in cardiac and skeletal muscles [82]. Accumulating evidence has shown that H19 gene polymorphisms are linked to cancer risk. Verhaegh et al. first reported that H19 rs2839698 variants significantly reduced the risk of bladder cancer among Caucasians, especially in non-muscle invasive bladder cancer [41]. Also, some studies have reported that the rs3741219 polymorphism was not associated with the risk of cancer, the rs2839698 polymorphism significantly increases the risk of gastrointestinal cancer, and the rs2107425 polymorphism had a protective effect on Caucasian population [81, 87, 88]. In order to accurately assess the association between H19 polymorphisms and the risk of cancer, we conducted a comprehensive analysis of all relevant potential studies.

Our findings suggested that the rs2839698, rs3741219 and rs3024270 polymorphisms, but not rs2107425, rs217727, or rs3741216 polymorphisms were associated with cancer risk in overall analysis. Among these, the rs2839698 polymorphism was dramatically related to increased cancer risk among Asians, while the rs210742 polymorphism was significantly associated with reduced cancer risk among Caucasians, indicating that ethnic differences in genetic backgrounds might influence the correlation. Using the RNA secondary structure prediction website, Gong et al. verified that the rs2107425 variant changed the minimum free energy of its centroid secondary structure and increased genetic susceptibility to cancer by impacting the H19 function and stability [51]. Further experimental functional studies are necessary to prove the exact mechanism. We found that rs2839698 SNP was positively associated with cancer susceptibility among Asians.

In the present study, the rs217727 mutation positively associated with increased risk of oral squamous cell carcinoma and lung cancer, but reduced the risk of hepatocellular carcinoma. Moreover, the rs2839698 polymorphism was significantly correlated with increased risk of gastric cancer, which was consistent with a previous study [88]. There were significant correlations between the rs2839698 polymorphism and cancer risk in hospital-based control, sample size and quality score subgroups. These results provided evidence that rs2839698 could modify cancer susceptibility based on ethnicity and cancer type. Furthermore, the discrepancy between our results and previous studies may be attributed to different genetic backgrounds and sample sizes. As for the rs3741219 polymorphism, a marginally notable correlation was discovered under recessive model in overall analysis. The positive results of higher quality score showed remarkable association with rs3024270 polymorphism. Moreover, we did not observe any relationships between rs3741216 rs3024270 and cancer in overall and subgroup analyses.

Among these H19 SNPs, rs217727, rs2839698, rs3741219, and rs3741216 located in exon region, as well as rs3024270 in intron region. SNPs at exon region are more likely to alter the H19 conserved folding structure or complementary sequences to target genes, and thus modify its binding affinity with interacting elements, while SNPs at intronic region may participate in alternative splicing and regulation of H19 transcript [86, 89]. Li et al. found that genetic variants of rs2839689, rs217727, rs2735971 and rs3024270 were closely associated with changes of H19 secondary structure in colorectal cancer [57]. It has been reported that the rs217727 polymorphism affected interactions between miRNAs and H19 and induced formation of target miRNA sites, such as hsa-miR-4804-5p and hsa-miR-8071, leading to the loss of hsa-miR-3960 and hsa-miR-8071 binding sites [73]. In addition, the rs2839698 mutation causes the loss of hsa-miR-24-1-5p and hsa-miR-24-2-5p, hsa-miR-566, and miR-675 [71, 75]. We speculated that the rs2839698 variation might hinder the binding of these targeted miRNA sites to the H19 3’-UTR, and then disrupt the reciprocal repression-regulatory loop between them, resulting in the tumorigenesis and progression.

Several limitations should be taken into account in the present study. First, heterogeneities were observed in most of the H19 SNPs, and subgroup analyses by source of control, cancer type, and ethnic diversity failed to completely eliminate these heterogeneities. Second, with regard to rs3741219, rs3024270 and rs3741216 polymorphism, all the included subjects were from Asian, and except for one study from Caucasians in rs217727 and rs2839698 polymorphism, other studies were involved with Asian population, which may not be applicable to other populations. Third, each type of cancer with only one study was assigned to subgroup analysis by other cancers, and the number of included studies for certain H19 polymorphisms was relatively limited in the subgroup analysis. Finally, due to the lack of available data on some factors such as alcohol consumption, smoking, lifestyle and effects of haplotype, we cannot evaluate the impact of gene-environmental and gene-gene interactions.

Conclusions

In conclusion, this meta-analysis demonstrated significant associations between H19 rs2839698 and rs3024270 and overall cancer risk. We found that H19 rs2107425 may be a protective factor for the Caucasian population, while rs2839698 may be a hazard factor for the Asian descent. Therefore, studies with larger sample sizes, diverse races and different cancer types are needed to further and better validate our findings.

Abbreviations: BC = breast cancer, LC = lung cancer, BLC = bladder cancer, GC = gastric cancer, CRC = colorectal cancer, PC = pancreatic cancer, OC = ovarian cancer, HCC: hepatocellular carcinoma, CC = cervical cancer, OSCC = oral squamous cell carcinoma, UCC = urothelial cell carcinoma, RCC = renal cell carcinoma, SNP = single nucleotide polymorphism, CI = confidence interval, HWE = Hardy-Weinberg equilibrium, NOS = Newcastle Ottawa Scale; OR = odds ratio.

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Wild CP. The global cancer burden: necessity is the mother of prevention. Nat Rev Cancer. 2019;19(3):123–4.

    Article  CAS  PubMed  Google Scholar 

  2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022, 72(1).

  3. Islami F, Goding Sauer A, Miller KD, Siegel RL, Fedewa SA, Jacobs EJ, McCullough ML, Patel AV, Ma J, Soerjomataram I, et al. Proportion and number of cancer cases and deaths attributable to potentially modifiable risk factors in the United States. CA Cancer J Clin. 2018;68(1):31–54.

    Article  PubMed  Google Scholar 

  4. Rossi M, Jahanzaib Anwar M, Usman A, Keshavarzian A, Bishehsari F. Colorectal Cancer and alcohol consumption-populations to molecules. Cancers (Basel) 2018, 10(2).

  5. Yan H, Ying Y, Xie H, Li J, Wang X, He L, Jin K, Tang J, Xu X, Zheng X. Secondhand smoking increases bladder cancer risk in nonsmoking population: a meta-analysis. Cancer Manag Res. 2018;10:3781–91.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Arshad R, Kiani MH, Rahdar A, Sargazi S, Barani M, Shojaei S, Bilal M, Kumar D, Pandey S. Nano-Based theranostic platforms for breast Cancer: a review of latest advancements. Bioeng (Basel) 2022, 9(7).

  7. Sargazi S, Er S, Mobashar A, Gelen SS, Rahdar A, Ebrahimi N, Hosseinikhah SM, Bilal M, Kyzas GZ. Aptamer-conjugated carbon-based nanomaterials for cancer and bacteria theranostics: a review. Chem Biol Interact. 2022;361:109964.

    Article  CAS  PubMed  Google Scholar 

  8. Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: new insights into immunomodulatory and nano-therapeutic approaches. J Control Release. 2022;349:844–75.

    Article  CAS  PubMed  Google Scholar 

  9. Janssen EM, Dy SM, Meara AS, Kneuertz PJ, Presley CJ, Bridges JFP. Analysis of patient preferences in Lung Cancer - estimating acceptable tradeoffs between Treatment Benefit and Side Effects. Patient Prefer Adherence. 2020;14:927–37.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Chen J, Jiang Y, Zhou J, Liu S, Qin N, Du J, Jin G, Hu Z, Ma H, Shen H, et al. Evaluation of CpG-SNPs in miRNA promoters and risk of breast cancer. Gene. 2018;651:1–8.

    Article  CAS  PubMed  Google Scholar 

  11. Zucman-Rossi J, Villanueva A, Nault J-C, Llovet JM. Genetic Landscape and biomarkers of Hepatocellular Carcinoma. Gastroenterology 2015, 149(5).

  12. Harati-Sadegh M, Sargazi S, Saravani M, Sheervalilou R, Mirinejad S, Saravani R. Relationship between miR-143/145 cluster variations and cancer risk: proof from a Meta-analysis. Nucleosides Nucleotides Nucleic Acids. 2021;40(5):578–91.

    Article  CAS  PubMed  Google Scholar 

  13. Sargazi S, Abghari AZ, Sarani H, Sheervalilou R, Mirinejad S, Saravani R, Eskandari E. Relationship between CASP9 and CASP10 gene polymorphisms and Cancer susceptibility: evidence from an updated Meta-analysis. Appl Biochem Biotechnol. 2021;193(12):4172–96.

    Article  CAS  PubMed  Google Scholar 

  14. Harati-Sadegh M, Mohammadoo-Khorasani M, Sargazi S, Saravani R, Shahraki S, Eskandari E. Quantitative Assessment of the Effects of IL-1ß -511 C > T variant on breast Cancer risk: an updated Meta-analysis of 3331 cases and 3609 controls. Lab Med. 2021;52(1):36–46.

    Article  PubMed  Google Scholar 

  15. Shastry BS. SNPs: impact on gene function and phenotype. Methods Mol Biol 2009, 578.

  16. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629–41.

    Article  CAS  PubMed  Google Scholar 

  17. Jalali S, Singh A, Maiti S, Scaria V. Genome-wide computational analysis of potential long noncoding RNA mediated DNA:DNA:RNA triplexes in the human genome. J Transl Med. 2017;15(1):186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen L, Zhang S. Long noncoding RNAs in cell differentiation and pluripotency. Cell Tissue Res. 2016;366(3):509–21.

    Article  CAS  PubMed  Google Scholar 

  19. Fatica A, Bozzoni I. Long non-coding RNAs: new players in cell differentiation and development. Nat Rev Genet 2014, 15(1).

  20. Tran N-T, Su H, Khodadadi-Jamayran A, Lin S, Zhang L, Zhou D, Pawlik KM, Townes TM, Chen Y, Mulloy JC, et al. The AS-RBM15 lncRNA enhances RBM15 protein translation during megakaryocyte differentiation. EMBO Rep. 2016;17(6):887–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bhan A, Mandal SS. Long noncoding RNAs: emerging stars in gene regulation, epigenetics and human disease. ChemMedChem. 2014;9(9):1932–56.

    Article  CAS  PubMed  Google Scholar 

  22. Taniue K, Akimitsu N. The functions and unique features of LncRNAs in Cancer Development and Tumorigenesis. Int J Mol Sci 2021, 22(2).

  23. Wilusz JE, Sunwoo H, Spector DL. Long noncoding RNAs: functional surprises from the RNA world. Genes Dev. 2009;23(13):1494–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang G, Lu X, Yuan L. LncRNA: a link between RNA and cancer. Biochim Biophys Acta. 2014;1839(11):1097–109.

    Article  CAS  PubMed  Google Scholar 

  25. Zou Y, Jiang Z, Yu X, Sun M, Zhang Y, Zuo Q, Zhou J, Yang N, Han P, Ge Z, et al. Upregulation of long noncoding RNA SPRY4-IT1 modulates proliferation, migration, apoptosis, and network formation in trophoblast cells HTR-8SV/neo. PLoS ONE. 2013;8(11):e79598.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shen Y, Xia E, Bhandari A, Wang X, Guo G. LncRNA PROX1-AS1 promotes proliferation, invasion, and migration in papillary thyroid carcinoma. Biosci Rep 2018, 38(5).

  27. Ou L, Wang D, Zhang H, Yu Q, Hua F. Decreased expression of mir-138-5p by lncRNA H19 in Cervical Cancer promotes Tumor Proliferation. Oncol Res. 2018;26(3):401–10.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Yang F, Bi J, Xue X, Zheng L, Zhi K, Hua J, Fang G. Up-regulated long non-coding RNA H19 contributes to proliferation of gastric cancer cells. FEBS J. 2012;279(17):3159–65.

    Article  CAS  PubMed  Google Scholar 

  29. Chi Y, Wang D, Wang J, Yu W, Yang J. Long non-coding RNA in the pathogenesis of cancers. Cells 2019, 8(9).

  30. Ghahramani Almanghadim H, Ghorbian S, Khademi NS, Soleymani Sadrabadi M, Jarrahi E, Nourollahzadeh Z, Dastani M, Shirvaliloo M, Sheervalilou R, Sargazi S. New Insights into the importance of long non-coding RNAs in Lung Cancer: future clinical approaches. DNA Cell Biol. 2021;40(12):1476–94.

    Article  CAS  PubMed  Google Scholar 

  31. Pachnis V, Brannan CI, Tilghman SM. The structure and expression of a novel gene activated in early mouse embryogenesis. EMBO J. 1988;7(3):673–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Kallen AN, Zhou X-B, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi J-S, Zhang H, et al. The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol Cell. 2013;52(1):101–12.

    Article  CAS  PubMed  Google Scholar 

  33. Koukoura O, Sifakis S, Zaravinos A, Apostolidou S, Jones A, Hajiioannou J, Widschwendter M, Spandidos DA. Hypomethylation along with increased H19 expression in placentas from pregnancies complicated with fetal growth restriction. Placenta. 2011;32(1):51–7.

    Article  CAS  PubMed  Google Scholar 

  34. Mutter GL, Stewart CL, Chaponot ML, Pomponio RJ. Oppositely imprinted genes H19 and insulin-like growth factor 2 are coexpressed in human androgenetic trophoblast. Am J Hum Genet. 1993;53(5):1096–102.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Brannan CI, Dees EC, Ingram RS, Tilghman SM. The product of the H19 gene may function as an RNA. Mol Cell Biol. 1990;10(1):28–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Matouk IJ, DeGroot N, Mezan S, Ayesh S, Abu-lail R, Hochberg A, Galun E. The H19 non-coding RNA is essential for human tumor growth. PLoS ONE. 2007;2(9):e845.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yoshimizu T, Miroglio A, Ripoche M-A, Gabory A, Vernucci M, Riccio A, Colnot S, Godard C, Terris B, Jammes H, et al. The H19 locus acts in vivo as a tumor suppressor. Proc Natl Acad Sci U S A. 2008;105(34):12417–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang L, Yang F, Yuan J-h, Yuan S-x, Zhou W-p, Huo X-s, Xu D, Bi H-s, Wang F. Sun S-h: epigenetic activation of the MiR-200 family contributes to H19-mediated metastasis suppression in hepatocellular carcinoma. Carcinogenesis. 2013;34(3):577–86.

    Article  PubMed  Google Scholar 

  39. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol. 2010;25(9):603–5.

    Article  PubMed  Google Scholar 

  40. 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(6):434–42.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Verhaegh GW, Verkleij L, Vermeulen SHHM, den Heijer M, Witjes JA, Kiemeney LA. Polymorphisms in the H19 gene and the risk of bladder cancer. Eur Urol. 2008;54(5):1118–26.

    Article  CAS  PubMed  Google Scholar 

  42. Song H, Ramus SJ, Kjaer SK, DiCioccio RA, Chenevix-Trench G, Pearce CL, Hogdall E, Whittemore AS, McGuire V, Hogdall C, et al. Association between invasive ovarian cancer susceptibility and 11 best candidate SNPs from breast cancer genome-wide association study. Hum Mol Genet. 2009;18(12):2297–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Quaye L, Tyrer J, Ramus SJ, Song H, Wozniak E, DiCioccio RA, McGuire V, Høgdall E, Høgdall C, Blaakaer J, et al. Association between common germline genetic variation in 94 candidate genes or regions and risks of invasive epithelial ovarian cancer. PLoS ONE. 2009;4(6):e5983.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Barnholtz-Sloan JS, Shetty PB, Guan X, Nyante SJ, Luo J, Brennan DJ, Millikan RC. FGFR2 and other loci identified in genome-wide association studies are associated with breast cancer in african-american and younger women. Carcinogenesis. 2010;31(8):1417–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Butt S, Harlid S, Borgquist S, Ivarsson M, Landberg G, Dillner J, Carlson J, Manjer J. Genetic predisposition, parity, age at first childbirth and risk for breast cancer. BMC Res Notes. 2012;5:414.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Yang C, Tang R, Ma X, Wang Y, Luo D, Xu Z, Zhu Y, Yang L. Tag SNPs in long non-coding RNA H19 contribute to susceptibility to gastric cancer in the chinese Han population. Oncotarget. 2015;6(17):15311–20.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Li S, Hua Y, Jin J, Wang H, Du M, Zhu L, Chu H, Zhang Z, Wang M. Association of genetic variants in lncRNA H19 with risk of colorectal cancer in a chinese population. Oncotarget. 2016;7(18):25470–7.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hua Q, Lv X, Gu X, Chen Y, Chu H, Du M, Gong W, Wang M, Zhang Z. Genetic variants in lncRNA H19 are associated with the risk of bladder cancer in a chinese population. Mutagenesis. 2016;31(5):531–8.

    Article  CAS  PubMed  Google Scholar 

  49. Xia Z, Yan R, Duan F, Song C, Wang P, Wang K. Genetic polymorphisms in long noncoding RNA H19 are Associated with susceptibility to breast Cancer in Chinese Population. Med (Baltim). 2016;95(7):e2771.

    Article  CAS  Google Scholar 

  50. Jin T, Wu X, Yang H, Liu M, He Y, He X, Shi X, Wang F, Du S, Ma Y, et al. Association of the mir-17-5p variants with susceptibility to cervical cancer in a chinese population. Oncotarget. 2016;7(47):76647–55.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Gong W-J, Peng J-B, Yin J-Y, Li X-P, Zheng W, Xiao L, Tan L-M, Xiao D, Chen Y-X, Li X, et al. Association between well-characterized lung cancer lncRNA polymorphisms and platinum-based chemotherapy toxicity in chinese patients with lung cancer. Acta Pharmacol Sin. 2017;38(4):581–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Guo QY, Wang H, Wang Y. LncRNA H19 polymorphisms associated with the risk of OSCC in chinese population. Eur Rev Med Pharmacol Sci. 2017;21(17):3770–4.

    PubMed  Google Scholar 

  53. Hassanzarei S, Hashemi M, Sattarifard H, Hashemi SM, Bahari G. Genetic polymorphisms in long noncoding RNA H19 are associated with breast cancer susceptibility in iranian population. Meta Gene. 2017;14:1–5.

    Article  Google Scholar 

  54. He TD, Xu D, Sui T, Zhu JK, Wei ZX, Wang YM. Association between H19 polymorphisms and osteosarcoma risk. Eur Rev Med Pharmacol Sci. 2017;21(17):3775–80.

    PubMed  Google Scholar 

  55. Hu P, Qiao O, Wang J, Li J, Jin H, Li Z, Jin Y. rs1859168 A > C polymorphism regulates HOTTIP expression and reduces risk of pancreatic cancer in a chinese population. World J Surg Oncol. 2017;15(1):155.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Lin Y, Fu F, Chen Y, Qiu W, Lin S, Yang P, Huang M, Wang C. Genetic variants in long noncoding RNA H19 contribute to the risk of breast cancer in a southeast China Han population. Onco Targets Ther. 2017;10:4369–78.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Li L, Guo G, Zhang H, Zhou B, Bai L, Chen H, Zhao Y, Yan Y. Association between H19 SNP rs217727 and lung cancer risk in a chinese population: a case control study. BMC Med Genet. 2018;19(1):136.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Yang M-L, Huang Z, Wang Q, Chen H-H, Ma S-N, Wu R, Cai W-S. The association of polymorphisms in lncRNA-H19 with hepatocellular cancer risk and prognosis. Biosci Rep 2018, 38(5).

  59. Yin Z, Cui Z, Li H, Li J, Zhou B. Polymorphisms in the H19 gene and the risk of lung Cancer among female never smokers in Shenyang, China. BMC Cancer. 2018;18(1):893.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yuan Z, Yu Y, Zhang B, Miao L, Wang L, Zhao K, Ji Y, Wang R, Ma H, Chen N, et al. Genetic variants in lncRNA H19 are associated with the risk of oral squamous cell carcinoma in a chinese population. Oncotarget. 2018;9(35):23915–22.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Cui P, Zhao Y, Chu X, He N, Zheng H, Han J, Song F, Chen K. SNP rs2071095 in LincRNA H19 is associated with breast cancer risk. Breast Cancer Res Treat. 2018;171(1):161–71.

    Article  CAS  PubMed  Google Scholar 

  62. Abdollahzadeh S, Ghorbian S. Association of the study between LncRNA-H19 gene polymorphisms with the risk of breast cancer. J Clin Lab Anal. 2019;33(3):e22826.

    Article  PubMed  Google Scholar 

  63. Hu C, Yang T, Pan J, Zhang J, Yang J, He J, Zou Y. Associations between H19 polymorphisms and neuroblastoma risk in chinese children. Biosci Rep 2019, 39(4).

  64. Li Z, Niu Y. Association between lncRNA H19 (rs217727, rs2735971 and rs3024270) polymorphisms and the risk of bladder cancer in chinese population. Minerva Urol Nefrol. 2019;71(2):161–7.

    Article  PubMed  Google Scholar 

  65. Safari MR, Mohammad Rezaei F, Dehghan A, Noroozi R, Taheri M, Ghafouri-Fard S. Genomic variants within the long non-coding RNA H19 confer risk of breast cancer in iranian population. Gene. 2019;701:121–4.

    Article  CAS  PubMed  Google Scholar 

  66. Wang G, Liu Q, Cui K, Ma A, Zhang H. Association between H19 polymorphisms and NSCLC risk in a Chinese Population. J BUON. 2019;24(3):913–7.

    PubMed  Google Scholar 

  67. Wu E-R, Chou Y-E, Liu Y-F, Hsueh K-C, Lee H-L, Yang S-F, Su S-C. Association of lncRNA H19 gene polymorphisms with the occurrence of Hepatocellular Carcinoma. Genes (Basel) 2019, 10(7).

  68. Wei M, Wang X, Luo B. Association between lncRNA H19 polymorphisms and susceptibility to gastric carcinoma and EBV-associated gastric carcinoma in han population in qingdao. Chin J Cancer Biotherapy. 2019;26(6):676–82.

    Google Scholar 

  69. Huang M-C, Chou Y-H, Shen H-P, Ng S-C, Lee Y-C, Sun Y-H, Hsu C-F, Yang S-F, Wang P-H. The clinicopathological characteristic associations of long non-coding RNA gene H19 polymorphisms with uterine cervical cancer. J Cancer. 2019;10(25):6191–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Yang P-J, Hsieh M-J, Hung T-W, Wang S-S, Chen S-C, Lee M-C, Yang S-F, Chou Y-E. Effects of Long Noncoding RNA H19 polymorphisms on Urothelial Cell Carcinoma Development. Int J Environ Res Public Health 2019, 16(8).

  71. Cao Q, Li P, Cao P, Qian J, Du M, Li L, Wang M, Qin C, Shao P, Zhang Z, et al. Genetic variant in long non-coding RNA H19 modulates its expression and predicts renal cell Carcinoma susceptibility and mortality. Front Oncol. 2020;10:785.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Ghapanchi J, Ranjbar Z, Mokhtari MJ, Koohpeima F, Derakhshan M, Khademi B, Ghaderi H, Sheikhbahaei S, Aliabadi E. The LncRNA H19 rs217727 polymorphism is Associated with oral squamous cell Carcinoma Susceptibility in Iranian Population. Biomed Res Int. 2020;2020:1634252.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Deng Y, Zhou L, Yao J, Liu Y, Zheng Y, Yang S, Wu Y, Li N, Xu P, Lyu L, et al. Associations of lncRNA H19 polymorphisms at MicroRNA binding Sites with Glioma susceptibility and prognosis. Mol Ther Nucleic Acids. 2020;20:86–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yu B, Chen J, Hou C, Zhang L, Jia J. LncRNA H19 gene rs2839698 polymorphism is associated with a decreased risk of colorectal cancer in a chinese Han population: a case-control study. J Clin Lab Anal. 2020;34(8):e23311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhang H-B, Zeng Y, Li T-L, Wang G. Correlation between polymorphisms in IGF2/H19 gene locus and epithelial ovarian cancer risk in chinese population. Genomics. 2020;112(3):2510–5.

    Article  CAS  PubMed  Google Scholar 

  76. Li W, Hua R-X, Wang M, Zhang D, Zhu J, Zhang S, Yang Y, Cheng J, Zhou H, Zhang J, et al. H19 gene polymorphisms and Wilms tumor risk in chinese children: a four-center case-control study. Mol Genet Genomic Med. 2021;9(2):e1584.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pei J-S, Chen C-C, Chang W-S, Wang Y-C, Chen J-C, Hsiau Y-C, Hsu P-C, Hsu Y-N, Tsai C-W. Bau D-T: significant Associations of lncRNA H19 genotypes with susceptibility to Childhood Leukemia in Taiwan. Pharmaceuticals (Basel) 2021, 14(3).

  78. Tan T, Li J, Wen Y, Zou Y, Yang J, Pan J, Hu C, Yao Y, Zhang J, Xin Y, et al. Association between lncRNA-H19 polymorphisms and hepatoblastoma risk in an ethic chinese population. J Cell Mol Med. 2021;25(2):742–50.

    Article  CAS  PubMed  Google Scholar 

  79. Zhang J, Liu M, Liang Y, Zhang M, Huang Z. Correlation between lncRNA H19 rs2839698 polymorphism and susceptibility to NK / T cell lymphoma in chinese population. J BUON. 2021;26(2):587–91.

    PubMed  Google Scholar 

  80. Khalil EH, Shaker OG, Hasona NA. Impact of rs2107425 polymorphism and expression of lncH19 and miR-200a on the susceptibility of Colorectal Cancer. Indian J Clin Biochem 2022.

  81. Li W, Jiang X, Jin X, Yan W, Liu Y, Li D, Zhao Z. Significant association between long non-coding RNA H19 polymorphisms and cancer susceptibility: a PRISMA-compliant meta-analysis and bioinformatics prediction. Med (Baltim). 2020;99(15):e19322.

    Article  CAS  Google Scholar 

  82. Berteaux N, Aptel N, Cathala G, Genton C, Coll J, Daccache A, Spruyt N, Hondermarck H, Dugimont T, Curgy J-J, et al. A novel H19 antisense RNA overexpressed in breast cancer contributes to paternal IGF2 expression. Mol Cell Biol. 2008;28(22):6731–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gao Y, Liu Y, Du L, Li J, Qu A, Zhang X, Wang L, Wang C. Down-regulation of mir-24-3p in colorectal cancer is associated with malignant behavior. Med Oncol. 2015;32(1):362.

    Article  PubMed  Google Scholar 

  84. Inoguchi S, Seki N, Chiyomaru T, Ishihara T, Matsushita R, Mataki H, Itesako T, Tatarano S, Yoshino H, Goto Y, et al. Tumour-suppressive microRNA-24-1 inhibits cancer cell proliferation through targeting FOXM1 in bladder cancer. FEBS Lett. 2014;588(17):3170–9.

    Article  CAS  PubMed  Google Scholar 

  85. Gao T, He B, Pan Y, Gu L, Chen L, Nie Z, Xu Y, Li R, Wang S. H19 DMR methylation correlates to the progression of esophageal squamous cell carcinoma through IGF2 imprinting pathway. Clin Transl Oncol. 2014;16(4):410–7.

    Article  CAS  PubMed  Google Scholar 

  86. McGeary SE, Lin KS, Shi CY, Pham TM, Bisaria N, Kelley GM, Bartel DP. The biochemical basis of microRNA targeting efficacy. Science 2019, 366(6472).

  87. Chu M, Yuan W, Wu S, Wang Z, Mao L, Tian T, Lu Y, Zhu B, Yang Y, Wang B, et al. Quantitative assessment of polymorphisms in H19 lncRNA and cancer risk: a meta-analysis of 13,392 cases and 18,893 controls. Oncotarget. 2016;7(48):78631–9.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Liu X, Zhao Y, Li Y, Zhang J. Quantitative assessment of lncRNA H19 polymorphisms and cancer risk: a meta-analysis based on 48,166 subjects. Artif Cells Nanomed Biotechnol. 2020;48(1):15–27.

    Article  CAS  PubMed  Google Scholar 

  89. Novikova IV, Hennelly SP, Sanbonmatsu KY. Structural architecture of the human long non-coding RNA, steroid receptor RNA activator. Nucleic Acids Res. 2012;40(11):5034–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

Wansheng Ji and Li Zhang conceptualized and designed the study, and proofread the final draft; Maoquan Yang and Mingwei Zhang searched the literature, extracted the data and prepared the final draft of manuscript. Qiong Wang, Xiaojing Guo, and Jinhua Gu conducted statistical analysis and prepared the figures. Maoquan Yang, and Peizhen Geng performed FPRP analysis and improved the introduction. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Wansheng Ji or Li Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Additional information

Publisher’s Note

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

Electronic supplementary material

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

Yang, M., Zhang, M., Wang, Q. et al. Six polymorphisms in the lncRNA H19 gene and the risk of cancer: a systematic review and meta-analysis. BMC Cancer 23, 688 (2023). https://doi.org/10.1186/s12885-023-11164-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-023-11164-y

Keywords