Skip to main content

Genetic variants of LncRNAs HOTTIP and MEG3 influence nasopharyngeal carcinoma susceptibility and clinicopathologic characteristics in the Southern Chinese population

Abstract

Objective

Recent studies have indicated that HOTTIP and MEG3 are associated with the initiation and progression of various types of tumors, including nasopharyngeal carcinoma (NPC). This investigation aimed to elucidate the impact of HOTTIP and MEG3 polymorphisms on the susceptibility and clinicopathologic characteristics of NPC.

Methods

This research employed next-generation sequencing and multiplex PCR to assess the polymorphisms of HOTTIP rs1859168 and MEG3 rs7158663 in 200 NPC and 200 healthy individuals respectively. HOTTIP and MEG3 expression were assessed via qRT-PCR assessment. Furthermore, the genotypes and alleles frequency of rs1859168 and rs7158663 were compared between healthy and NPC individuals to elucidate their influence on NPC susceptibility and relation with clinicopathologic characteristics.

Results

In comparison with the healthy cohort, the presence of HOTTIP rs1859168 CC genotype and the C allele were markedly linked with increased NPC incidence (p < 0.05). Furthermore, the MEG3 rs7158663 AA genotype and the A allele also indicated an increased risk of NPC (p < 0.05). The subgroup analysis of age, EBV infection, gender, nationality, smoking, and drinking status revealed no marked association between rs1859168 and rs7158663 genotypes and these potential confounding factors. Moreover, it was observed that rs1859168 CC and rs7158663 AA genotypes were related to local tumor invasion and lymph node metastasis. Additionally, HOTTIP indicated a marked elevation, while MEG3 substantially reduced in NPC samples than the normal nasopharyngeal biospecimens. Patients who carried CC or CA genotypes rather than the HOTTIP rs1859168 AA genotype, had substantially higher HOTTIP levels, while patients with rs7158663 AA or GA genotypes indicated notably lower expression of MEG3 than GG genotype carriers.

Conclusion

Individuals with genetic variants of HOTTIP rs1859168 and MEG3 rs7158663 might have an increased risk of NPC susceptibility and related clinicopathologic characteristics, potentially by affecting the expression of HOTTIP and MEG3.

Introduction

Nasopharyngeal carcinoma (NPC) is a malignant tumor that is distributed in various geographical and ethnic regions and has an increased incidence rate in Southern China [1]. It has a complex etiology, however, infection by Epstein-Barr virus (EBV) is the primary cause. EBV transforms normal host cells into cancerous types and can stimulate or inhibit various host cellular processes and mechanisms. Furthermore, NPC is not only influenced by genetics, but also environmental risk factors, including prolonged cigarette smoking, drinking, and various dietary factors. However, different individuals with the same environmental risk factors present distinct characteristics, indicating that in NPC formation, genetic susceptibility is the primary risk factor. Single nucleotide polymorphism (SNP) is the most common type of heritable variation in humans, which can affect individual susceptibility to tumors and is essentially linked to tumor pathogenesis, advancement, and prognosis [2].

Long non-coding RNAs (lncRNAs) comprise more than 200 nucleotides and regulate various cellular processes at the post-transcriptional, epigenetic, and transcriptional levels. Recently, it has been indicated that abnormal lncRNA levels are linked with the initiation, metastasis, susceptibility, and prognosis of multiple human cancers, such as gastric cancer, breast cancer, and NPC [3,4,5]. According to GWAS, only 7% of disease-related loci are present in protein-coding regions, while 93% are in non-coding regions, indicating that genetic alterations in non-coding sites may play a crucial part in regulating disease progression [6]. Furthermore, the literature has revealed that SNPs in lncRNA genes can influence the lncRNA expression, subsequently affecting their function or structure, thereby impacting tumorigenesis and cancer prognosis [7,8,9].

HOTTIP is a lncRNA that is transcribed from the 5’ end of HOXA cluster, involved in the HOX gene network, and can influence cell function by targeting HOXA gene transcription, resulting in epigenetic alteration and promoting tumor occurrence and progression [10]. HOTTIP is considered an oncogene as its overexpression has been observed in different cancers, such as hepatocellular carcinoma (HCC) [11], pancreatic ductal adenocarcinoma [12], gastric cancer [13], and NPC [14].

Maternally expressed gene 3 (MEG3) is an imprinted gene, which encodes about 1.6 kb long non-coding RNA and has been indicated as a tumor suppressor. Much research has indicated that MEG3 expression is inhibited in different tumors, such as gastric [15], ovarian [16], and prostate cancers [17], as well as NPC [18]. Moreover, it has been revealed that HOTTIP up-regulation or MEG3 down-regulation is associated with cancer pathogenesis and progression.

Some studies have indicated the association of HOTTIP and MEG3 genetic polymorphisms with cancer susceptibility. For instance, Abdelaleem et al. revealed that HOTTIP rs1859168 variation was linked with an increased susceptibility to breast cancer (BC) and might be utilized as an indicator and therapeutic target for BC patients [19]. Wang et al. demonstrated that the rs3807598 and rs2067087 variants of HOTTIP were correlated with elevated gastric cancer risk [20]. Furthermore, Elhelaly revealed that MEG3 rs7158663 was linked with colorectal cancer (CRC) risk and therefore may serve as its diagnostic and prognostic index [21]. Mohammed et al. found that MEG3 rs7158663 promotes the risk of HCC and may be poor diagnostic and prognostic factors for HCC patients [22]. However, the associations of HOTTIP and MEG3 SNPs with NPC remain unclear. This investigation aimed to elucidate if rs1859168 and rs7158663 genetic variants are linked with increased NPC susceptibility or development and assess whether these polymorphisms could affect HOTTIP and MEG3 expressions and their possible correlations with the clinicopathological features.

Materials and methods

Enrolled participants

This research comprised 200 newly diagnosed NPC patients who were pathologically confirmed at the Department of Otolaryngology of Minzu Hospital of Guangxi Zhuang Autonomous Region (Guangxi, China) from January 2018 and December 2022. Additionally, 200 healthy individuals were also recruited as healthy controls from the physical examination center of the same hospital. Patients who underwent chemotherapy or radiotherapy before surgery or had other types of cancer were not selected in the NPC cohort. Furthermore, for the healthy cohort, individuals with cancer history or those who were related to the enrolled patients were excluded. Both the cohorts were sex, age, and residential area matched. Moreover, 48 NPC tumor tissues and 20 normal nasopharyngeal biospecimens were collected for RNA expression analysis. This investigation was authorized by the Ethics Committee of Minzu Hospital of Guangxi Zhuang Autonomous Region and signed consent was acquired from each participant before the study began.

DNA isolation and genotyping

The acquired tissues and venous blood were kept at -80℃ before experiments. HOTTIP rs1859168 and MEG3 rs7158663 polymorphisms for both tissue and blood samples were identified via multiplex PCR and next-generation sequencing. Briefly, DNA was isolated from the samples via the TIANamp Genomic DNA Kit (Tiangen Biotech, Beijing, China), per the kit’s guide. The acquired DNA was then quantified, and purified and its integrity was assessed using Nanodrop one ultra-micro-spectrophotometer and agarose gel electrophoresis. Subsequently, PCR was carried out using primers synthesized by Qike Biotechnology (Beijing, China). Table 1 indicates the optimized reaction conditions. Sequencing and genotyping were conducted by Qike Biotechnology (Guangzhou, China) after electrophoretic confirmation of the sequencing products. The genotypes of HOTTIP rs1859168 were designated as A/A, A/C, or C/C, while MEG3 rs7158663 were designated as A/A, A/G, or G/G.

Table 1 Primer sequence and the reaction condition for HOTTIP rs1859168 and MEG3 rs7158663

Quantitative real-time polymerase chain reaction (qRT-PCR)

Whole tissue RNA was acquired via the TRIzol reagent kit (Tiangen Biotech, Beijing, China), which was then quantified and purified before assessing its integrity using a Nanodrop one ultra-micro-spectrophotometer and agarose gel electrophoresis. Afterward, cDNA was prepared using the RevertAid cDNA Synthesis Kit (Thermo Fisher, Beijing, China), per the kit’s guide. Subsequently, an Applied Biosystems 7500 Real-Time PCR system was employed for qRT-PCR analysis. GAPDH was selected as endogenous control and the 2-ΔΔCt algorithm was applied to quantify HOTTIP and MEG3 expressions.

The primers synthesized by Sangon Biotechnology (Shanghai, China) were as follows:

HOTTIP:

Forward: 5’-CCTAAAGCCACGCTTCTTTG-3’.

Reverse: 5’-TGCAGGCTGGAGATCCTACT-3’.

MEG3:

Forward: 5’-GGGAAGGGACCTCGAATGTG − 3’.

Reverse: 5’-CTGTCCCGTGGGAATAGGTG-3’.

GAPDH:

Forward: 5’-GTCAAGGCTGAGAACGGGAA-3’.

Reverse: 5’-AAATGAGCCCCAGCCTTCTC-3’.

Statistical analysis

For statistical assessment, SPSS software v20.0 was utilized. The distribution frequencies of genotype and allele were compared and Hardy-Weinberg equilibrium (HWE) was elucidated by Chi-squared test. The relative risk related to various genotypes and alleles was elucidated by logistic regression analyses based on 95% confidence intervals (95% CIs) and calculated odds ratios (ORs). Furthermore, a non-parametric Mann-Whitney U test was carried out to compare the relative expressions of HOTTIP and MEG3 in tissue samples of different genotypic groups. A p-value < 0.05 was deemed statistically significant.

Results

Clinical, demographic, and pathological features of the participants

Table 2 summarizes the comprehensive data on the NPC cases and control cohort. No marked differences in age, nationality, smoking, gender, and drinking proportion were identified between NPC patients and healthy individuals (p > 0.05). However, the EBV-DNA positive rate was substantially higher in NPC patients than in the controls (p < 0.001).

Table 2 Characteristics of the participants

Distribution frequency of the genotypes and alleles of rs7158663 and rs1859168 in NPC patients and healthy participants

Figures 1 and 2 present the genotyping sequencing data of rs1859168 and rs7158663. The distribution of these genotypes in patients and controls conformed to the Hardy-Weinberg equilibrium (all p > 0.05). Additionally, for the same cases, the genotypes of tissues were consistent with those of blood samples. The distribution of various alleles and genotypes of rs1859168 and rs7158663 are presented in Table 3.

The HOTTIP rs1859168 CC genotype was markedly linked with increased NPC incidence than the AA genotype (95% CI: 1.511–4.890, OR: 2.718, p = 0.001). Furthermore, the dominant model indicated a notable association of the AC + CC genotypes with increased NPC susceptibility (95% CI: 1.198–2.911, OR: 1.867, p = 0.005). Additionally, the recessive model revealed that the CC genotype was linked with a higher risk of NPC (95% CI: 1.201–3.284, OR: 1.991, p = 0.006). Moreover, the C allele was also related to an elevated risk of NPC (95% CI: 1.215–2.218, OR: 1.608, p = 0.001).

Compared with the MEG3 rs7158663 GG genotype, the AA genotype demonstrated substantial association with enhanced risk of NPC (95% CI: 1.194–4.007, OR: 2.187, p = 0.010). Furthermore, the GA + AA genotypes in the dominant model were notably linked with an enhanced NPC susceptibility (95% CI: 1.028–2.262, OR: 1.525, p = 0.036). Meanwhile, in the recessive model, the AA genotype showed a relation with increased NPC risk (95% CI: 1.088–3.442, OR: 1.935, p = 0.023). Moreover, the A allele was also linked with increased NPC risk (95% CI: 1.132–2.051, OR: 1.524, p = 0.005).

Fig. 1
figure 1

Genotype sequencing map of the HOTTIP rs1859168 polymorphism. (A) rs1859168- A/A; (B) rs1859168-A/C; (C) rs1859168-C/C

Fig. 2
figure 2

Genotype sequencing map of the MEG3 rs7158663 polymorphism. (A) rs7158663- G/G; (B) rs7158663-G/A; (C) rs7158663-A/A

Table 3 Distribution frequencies of allele and genotype in patients and controls

Stratified analyses of genotype distributions by gender, age, EBV-DNA, nationality, status of smoking and drinking in NPC patients

Further stratified analysis indicated that gender, EBV infection, age, smoking, nationality, and drinking status do not affect the association of HOTTIP rs1859168 and MEG3 rs7158663 with NPC susceptibility (all p > 0.05) (Table 4). These datas suggest that potential confounding variables had few modification effects on NPC susceptibility related to rs1859168 and rs7158663 genotypes.

Table 4 Stratification analyses of rs1859168 and rs7158663 genotypes among NPC patients

Associations of rs1859168 and rs7158663 genotypes with clinicopathological features of NPC patients

The association of different HOTTIP rs1859168 and MEG3 rs7158663 genotypes with the NPC clinicopathological features was assessed (Table 5).

It was revealed that in comparison with the HOTTIP rs1859168 AA genotype, patients carrying the CC genotype had a greater incidence of III + IV clinical stage (OR: 2.311, p = 0.044), T3 + T4 local tumor invasion (OR: 2.575, p = 0.027), N2 + N3 lymph node status (OR: 2.914, p = 0.017).

Furthermore, patients carrying MEG3 rs7158663 AA genotype had an increased risk of T3 + T4 local tumor invasion (OR: 2.771, p = 0.011) and N2 + N3 lymph node status (OR: 2.511, p = 0.023) than those with GG genotype.

Table 5 Correlation of rs7158663 and rs1859168 genotypes with clinicopathologic features of NPC

HOTTIP and MEG3 expression levels in NPC patients with different clinicopathological features

To understand how HOTTIP and MEG3 are related to NPC occurrence and progression, their expression levels in NPC tissues and their association with different clinicopathological parameters were assessed. It was revealed that HOTTIP had a higher expression in NPC tissues than control nasopharyngeal biospecimens (p < 0.001). Furthermore, the expression of HOTTIP was markedly elevated in NPC tissues with III + IV clinical stage, T3 + T4 local tumor invasion, and N2 + N3 lymph node status than those with I + II clinical stage, T1 + T2 local tumor invasion, and N0 + N1 lymph node status (all p < 0.05) (Fig. 3). Conversely, MEG3 indicated reduced expression in NPC patients than in healthy nasopharyngeal biospecimens (p < 0.001). Moreover, MEG3 indicated notably reduced expression in NPC tissues with III + IV clinical stage, T3 + T4 local tumor invasion, and N2 + N3 lymph node status than those with I + II clinical stage, T1 + T2 local tumor invasion, and N0 + N1 lymph node status (all p < 0.05) (Fig. 4).

Fig. 3
figure 3

(A) The relative expression of HOTTIP in NPC tumor tissues and normal nasopharyngeal biospecimens. (B) The relative expression of HOTTIP in different clinical stages, (C) local tumor invasions, and (D) lymph node status

Fig. 4
figure 4

(A) The relative expression of MEG3 in NPC tumor tissues and normal nasopharyngeal biospecimens. (B) The relative expression of MEG3 in different clinical stages, (C) local tumor invasions, and (D) lymph node status

The effect of rs1859168 and rs7158663 on HOTTIP and MEG3 expressions in NPC

Subsequently, the association of different HOTTIP rs1859168 or MEG3 rs7158663 genotypes with HOTTIP or MEG3 expression in NPC patients was assessed.

It was revealed that HOTTIP had a higher expression of different genotypes in NPC tissues than control nasopharyngeal biospecimens (Fig. 5A). Moreover, in comparison with the HOTTIP rs1859168 AA genotype, NPC patients carrying AC or CC genotypes exhibited notably higher levels of HOTTIP (Fig. 5B).

Furthermore, It was revealed that MEG3 had a lower expression of different genotypes in NPC tissues than control nasopharyngeal biospecimens (Fig. 6A). NPC patients carrying the rs7158663 GA or AA genotypes indicated markedly reduced levels of MEG3 than those with the GG genotype (Fig. 6B).

Fig. 5
figure 5

(A) The relative expression of HOTTIP of different genotypes in NPC tumor tissues and normal nasopharyngeal biospecimens. (B) The relative expression of HOTTIP of different genotypes in NPC tumor tissues

Fig. 6
figure 6

(A) The relative expression of MEG3 of different genotypes in NPC tumor tissues and normal nasopharyngeal biospecimens. (B) The relative expression of MEG3 of different genotypes in NPC tumor tissues

Discussion

Nasopharyngeal carcinoma originates from the nasopharyngeal epithelium and is an aggressive type of neck and head cancer. Because of the narrow nasopharyngeal cavity, hidden onset, and deep tissue structure, at the time of diagnosis > 70% of patients have reached locoregionally advanced stages [23]. Therefore, identification of new genetic bio-indices and therapeutic targets is essential for early diagnosis and treatment. The research suggests that lncRNAs can serve as genetic bio-indices of NPC [24]. Furthermore, SNPs in lncRNA genes have been indicated to influence NPC susceptibility and progression [25, 26]. However, the association of HOTTIP rs1859168 and MEG3 rs7158663 polymorphism with NPC susceptibility and tumor progression remains undetermined. This current study is the first to indicate that the HOTTIP rs1859168 CC and MEG3 rs7158663 AA genotypes are markedly correlated with increased susceptibility to NPC and clinical pathological features. Moreover, these genotypes also influence HOTTIP and MEG3 expression in NPC patients.

HOTTIP is an antisense non-coding transcript located at the 5′end of the HOXA gene cluster and is considered an oncogene, valuable biomarker, and therapeutic target in various malignancies [11,12,13]. It also modulates the expression of cancer-associated genes at both transcriptional and post-transcriptional levels. For example, Lin et al. revealed that HOTTIP regulated HOXA13 gene transcription in the cells of esophageal squamous cell carcinoma, thereby influencing its tumorigenesis and metastasis [27]. Han et al. found that HOTTIP directly bonds to miR-148a-3p to increase WNT1 expression, facilitating BC stemness [28]. In addition, HOTTIP alters immune cell’s anti-cancer effects, thereby enhancing their immune evasion. For example, it has been observed to enhance IL-6 expression in ovarian cancer, consequently up-regulating PD-L1 expression in neutrophils, thereby accelerating tumor immune escape [29]. Two researches have elucidated the link between HOTTIP and NPC. Feng et al. reported that HOTTIP expression was markedly increased in NPC cells and tissues, and it promoted tumorigenesis by modulating the HOXA13 expression [30]. Shen et al. revealed that HOTTIP levels were markedly enhanced in NPC tissues and it promoted NPC cell’s ability to proliferate, migrate, and invade by sponging miR-4301 [14]. In the current study, it was revealed that HOTTIP expression was notably elevated in NPC tissues and it was positively related to local tumor invasion, clinical stage, and lymph node metastasis. Based on these results, HOTTIP might have a tumor-promoting role in NPC.

Many recent studies have indicated the associations of HOTTIP variants with cancer risk. Furthermore, these variants influenced HOTTIP expression by influencing the binding sites of transcription factors. According to Wang et al., HOTTIP rs2067087 and rs3807598 were related to increased risk of gastric cancer by affecting HOTTIP expression [20]. Another report revealed that the HOTAIR SNPs and their interactions with the HOTTIP rs1859168 polymorphism significantly increased the risk of gastric cancer [31]. Abdelaleem et al. suggested that individuals carrying the rs1859168 CC genotype had an elevated risk of BC as they influence HOTTIP and miR-615-3p levels [19]. Ali et al. demonstrated that rs1859168 C allele and CC genotype were associated with an elevated risk of CRC by up-regulating HOTTIP expression in the Egyptian population [32]. On the contrary, Duan et al. indicated that rs1859168 was markedly linked with an alleviated risk of gastric cancer in the Chinese population [33]. Moreover, according to Hu et al., the rs1859168 C allele and CC genotype notably reduced the susceptibility to pancreatic cancer by down-regulating the HOTTIP expression [34]. Thus, the tumor type and study population might be the primary factor influencing rs1859168 as a susceptibility marker for tumors. Here, the association of rs1859168 polymorphism with NPC risk was evaluated, which indicated that rs1859168 C allele and CC genotype notably increased NPC risk. Furthermore, the genotype-phenotype analysis showed that rs1859168 CC or AC genotype was related to the expression of HOTTIP, indicating that this variant might impact the HOTTIP expression by modulating its transcript function, thereby correlating with NPC risk.

It has been reported that MEG3 levels are reduced in many human malignancies, and it functions as a tumor suppressor [15,16,17], by modulating common cell signal transduction pathways. Yan et al. indicated that in retinoblastoma, MEG3 overexpression enhanced cell apoptosis and decreased cell proliferation and migration via PI3K/Akt/mTOR pathway inactivating [35]. Lv et al. exhibited that in non-small cell lung cancer, high MEG3 levels elevated miR-21-5p and PTEN expressions and inhibited cellular migration and invasion via the miR-21 5p/PTEN axis [36]. In most tumor types, MEG3 acts as a tumor suppressor by directly interacting with miRNAs or acting as their molecular sponge. For instance, Tao et al. revealed that MEG3 overexpression suppressed cell proliferation and invasion while stimulating apoptosis in ovarian cancer by sponging miR-205-5p [37]. According to Shan et al., MEG3 was notably reduced in bladder cancer cells, and it can alleviate the progression of bladder cancer by modulating PTEN and miR-494 [38]. Recently, Lin et al. demonstrated that in NPC cells, MEG3 levels were reduced, stimulating NPC cells’ autophagy and apoptosis by interacting with miR-21 to elevate PTEN expression [18]. Moreover, Zhou et al. proved that MEG3 was downregulated in NPC cells and tissues and its overexpression inhibited invasion, epithelial-mesenchymal transition, and migration of NPC cells by modulating SQSTM1 [39]. In accordance with the evidence mentioned previously, this current study was also found that MEG3 reduced notably in NPC tissues relative to normal nasopharyngeal biospecimens. In addition, MEG3 expression was negatively related to local tumor invasion, clinical stage, and lymph node metastasis in NPC.

Much evidence has indicated that MEG3 polymorphisms are linked with cancer susceptibility. rs7158663 is present on the MEG3 transcript and is the most interesting polymorphic locus. Bioinformatics assessments have indicated that rs7158663 polymorphism can alter the folding structure of local RNA and affect the interactions of miRNA-lncRNA, thereby affecting the levels of MEG3. Various research has elucidated the link between this polymerphism and cancer susceptibility. For example, Elhelaly et al. suggested that the rs7158663 AA genotype was related to CRC risk in Egyptian patients and might be utilized as a diagnostic and prognostic marker for CRC patients [21]. Gao et al. found that the rs7158663 GA or AA genotype was linked with an elevated risk of CRC in the Chinese population [40]. Per Kong et al., MEG3 rs7158663 AA or GA genotype and A allele carriers could substantially elevate gastric cancer risk [41], while according to Olfat et al., these carriers have increased susceptibility to BC [42]. However, some research indicated that the rs7158663 variant was not linked with cancer risk. Zhuo et al. revealed that the rs7158663 polymorphism was not related to neuroblastoma risk [43], while Yang et al. indicated that it was not linked with lung cancer susceptibility [44]. These inconsistent results may differ from tumor type and the study population. This is the first research to evaluate the association of MEG3 polymorphism with NPC, which revealed that the rs7158663 AA genotype was linked with an elevated risk of NPC, and the carriers of GA or AA genotypes had markedly reduced levels of MEG3. Therefore, it was speculated that the polymorphism of rs7158663 might affect an individual’s susceptibility to NPC by regulating MEG3 expression.

In addition, it has been indicated that SNPs in lncRNA genes influence the function of these lncRNAs and promote the progression of various tumors [45,46,47]. Few studies have provided evidence supporting the link between the HOTTIP rs1859168 and MEG3 rs7158663 genetic variants and the progression of tumors. Abdelaleem et al. revealed that in BC, the rs1859168 CC genotype and C allele were markedly linked with higher TNM staging [19]. Ali et al. also suggested that HOTTIP rs1859168 was substantially related to lymph node metastasis, distant metastasis, and grade III of CRC [32]. They also indicated that MEG3 rs7158663 polymorphism was notably linked with higher TNM staging and larger tumor size of BC in the Egyptian population [48]. Mohammed et al. verified that in HCC, the rs7158663 variant was markedly correlated with larger tumor size and advanced stage [22]. Here, the association of HOTTIP rs1859168 and MEG3 rs7158663 genetic variants with clinicopathological features of NPC patients was assessed. Patients carrying rs1859168 CC or rs7158663 AA genotypes had an increased risk of local tumor invasion and lymph node metastasis than rs1859168 AA or rs7158663 GG genotype carriers. These data suggests that HOTTIP rs1859168 and MEG3 rs7158663 variants might be associated with the NPC progression.

Limitations

The limitations of this study include: (1) It is a case-control study, therefore is subjected to inevitable selection bias. (2) This investigation was executed as a single-center and requires further large-scale multicenter research to validate its data. (3) The molecular mechanisms of how rs1859168 and rs7158663 variants influence NPC pathogenesis and the link of these polymorphisms with HOTTIP and MEG3 expression in other ethnic groups need further elucidation.

Conclusion

Altogether, it was revealed that HOTTIP rs1859168 and MEG3 rs7158663 genetic variants were related to an increased risk of NPC. Furthermore, rs1859168 CC or rs7158663 AA genotypes were associated with NPC patients’ clinicopathologic characteristics. Moreover, it was verified that the rs1859168 CC/AC or rs7158663 AA/GA genotypes were linked to high HOTTIP or low MEG3 levels in NPC patients. Comprehensive mechanisms of rs1859168 and rs7158663 variants influencing NPC pathogenesis warrant further research.

Data availability

The raw data supporting the results and conclusions of this article will be made available by the authors. All data generated or analyzed presented in the study are included in the article.

References

  1. Zhang Y, Chen L, Hu GQ, Zhang N, Zhu XD, Yang KY, et al. Gemcitabine and Cisplatin Induction Chemotherapy in nasopharyngeal carcinoma. N Engl J Med. 2019;381(12):1124–35.

    Article  CAS  PubMed  Google Scholar 

  2. Olbromski PJ, Bogacz A, Bukowska M, Kamiński A, Moszyński R, Pawlik P, et al. Analysis of the polymorphisms and expression levels of the BCL2, BAX and c-MYC genes in patients with ovarian Cancer. Int J Mol Sci. 2023;24(22):null.

    Article  Google Scholar 

  3. Li X, Du Y, Wang Y. The value of LncRNA SNHG5 as a marker for the diagnosis and prognosis of gastric cancer. Am J Translational Res. 2021;13(5):5420–7.

    CAS  Google Scholar 

  4. Zong S, Dai W, Guo X, Wang K. LncRNA-SNHG1 promotes macrophage M2-like polarization and contributes to breast cancer growth and metastasis. Aging. 2021;13(19):23169–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Yang H, Pan Y, Zhang J, Jin L, Zhang X. LncRNA FOXD3-AS1 Promotes the Malignant Progression of Nasopharyngeal Carcinoma Through Enhancing the Transcription of YBX1 by H3K27Ac Modification. Frontiers in oncology. 2021;11(null):715635.

  6. Freedman ML, Monteiro AN, Gayther SA, Coetzee GA, Risch A, Plass C, et al. Principles for the post-GWAS functional characterization of cancer risk loci. Nat Genet. 2011;43(6):513–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sun YH, Chou YH, Tsai HY, Hsiao YH, Lee CY, Yang SF, et al. Impact of genetic variants of long noncoding RNA metastasis-Associated Lung Adenocarcinoma transcript 1 on uterine cervical Cancer. J Cancer. 2022;13(7):2150–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yeh JC, Chen YT, Chou YE, Su SC, Chang LC, Chen YL, et al. Interactive effects of CDKN2B-AS1 gene polymorphism and habitual risk factors on oral cancer. J Cell Mol Med. 2023;27(21):3395–403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Xiang X, Chen L, He J, Ma G, Li Y. LncRNA GAS5 rs145204276 polymorphism reduces renal cell Carcinoma susceptibility in Southern Chinese Population. J Inflamm Res. 2022;15(null):1147–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ghafouri-Fard S, Dashti S, Taheri M. The HOTTIP (HOXA transcript at the distal tip) lncRNA: Review of oncogenic roles in human. Biomedicine & pharmacotherapy. 2020;127(null):110158.

  11. Wu L, Yang Z, Zhang J, Xie H, Zhou L, Zheng S. Long noncoding RNA HOTTIP expression predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Hepatobiliary Surg Nutr. 2018;7(6):429–39.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Wong CH, Li CH, He Q, Chan SL, Tong JH, To KF, et al. Ectopic HOTTIP expression induces noncanonical transactivation pathways to promote growth and invasiveness in pancreatic ductal adenocarcinoma. Cancer Lett. 2020;477(null):1–9.

    Article  CAS  PubMed  Google Scholar 

  13. Xin Y, Li X, Zhang M, Shang Z, Luo Z, Wang Y, et al. Fusobacterium nucleatum-induced exosomal HOTTIP promotes gastric cancer progression through the microRNA-885-3p/EphB2 axis. Cancer Sci. 2023;114(6):2360–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Shen M, Li M, Liu J, Long Noncoding RNAHOTTIP. Promotes nasopharyngeal Cancer Cell Proliferation, Migration, and Invasion by inhibiting miR-4301. Med Sci Monitor: Int Med J Experimental Clin Res. 2019;25(null):778–85.

    Article  CAS  Google Scholar 

  15. Zhang Y, Wang Y, Jiang Y, Bai H, Wen Y. The meaningful function of the emerging clinical Targets-lncRNA MEG3 in gastric Cancer. Curr Pharm Design. 2023;29(28):2204–12.

    Article  CAS  Google Scholar 

  16. Li Y, Zhang L, Zhao Y, Peng H, Zhang N, Bai W. MEG3 sponges miRNA-376a and YBX1 to regulate angiogenesis in ovarian cancer endothelial cells. Heliyon. 2023;9(2):e13204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhou Y, Yang H, Xia W, Cui L, Xu R, Lu H, et al. LncRNA MEG3 inhibits the progression of prostate cancer by facilitating H3K27 trimethylation of EN2 through binding to EZH2. J BioChem. 2020;167(3):295–301.

    Article  CAS  PubMed  Google Scholar 

  18. Lin L, Liu X, Lv B. Long non-coding RNA MEG3 promotes autophagy and apoptosis of nasopharyngeal carcinoma cells via PTEN up-regulation by binding to microRNA-21. J Cell Mol Med. 2021;25(1):61–72.

    Article  CAS  PubMed  Google Scholar 

  19. Abdelaleem OO, Shaker OG, AbdelHafez MN, Abdelghaffar NK, Eid HM, Zaidan M, et al. The influence of rs1859168 polymorphism on serum expression of HOTTIP and its target mir-615-3p in Egyptian patients with breast Cancer. Biomolecules. 2021;11(5):null.

    Article  Google Scholar 

  20. Wang BG, Li YZ, Ding HX, Lv Z, Xu Q, Yuan Y. HOTTIP polymorphism may affect gastric cancer susceptibility by altering HOTTIP expression. Biosci Rep. 2020;40(8):null.

    Article  Google Scholar 

  21. Elhelaly Elsherbeny M, Ramadan Elsergany A, Gamil Shaker O. Association of lncRNA MEG3 Rs7158663 polymorphism and serum expression with colorectal Cancer in Egyptian patients. Rep Biochem Mol Biology. 2023;12(1):102–11.

    Google Scholar 

  22. Mohammed SR, Shaker OG, Mohamed MM, Abdelhafez Mostafa MN, Gaber SN, Ali DY, et al. The emerging role of lncRNA MEG3 and MEG3 rs7158663 in hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 2022;26(1):11–21.

    CAS  PubMed  Google Scholar 

  23. Zhang Y, Sun Y, Ma J. Induction gemcitabine and cisplatin in locoregionally advanced nasopharyngeal carcinoma. Cancer Commun (London England). 2019;39(1):39.

    Google Scholar 

  24. Liang YL, Zhang Y, Tan XR, Qiao H, Liu SR, Tang LL, et al. A lncRNA signature associated with tumor immune heterogeneity predicts distant metastasis in locoregionally advanced nasopharyngeal carcinoma. Nat Commun. 2022;13(1):2996.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Guo Z, Wang YJ, He BS, Zhou J. Linc00312 single nucleotide polymorphism as Biomarker for Chemoradiotherapy Induced Hematotoxicity in Nasopharyngeal Carcinoma patients. Dis Markers. 2022;2022(null):6707821.

    Google Scholar 

  26. Guo Z, Li Z, Zhang M, Bao M, He B, Zhou X. LncRNA FAS-AS1 upregulated by its genetic variation rs6586163 promotes cell apoptosis in nasopharyngeal carcinoma through regulating mitochondria function and Fas splicing. Sci Rep. 2023;13(1):8218.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lin C, Wang Y, Wang Y, Zhang S, Yu L, Guo C, et al. Transcriptional and posttranscriptional regulation of HOXA13 by lncRNA HOTTIP facilitates tumorigenesis and metastasis in esophageal squamous carcinoma cells. Oncogene. 2017;36(38):5392–406.

    Article  CAS  PubMed  Google Scholar 

  28. Han L, Yan Y, Zhao L, Liu Y, Lv X, Zhang L, et al. LncRNA HOTTIP facilitates the stemness of breast cancer via regulation of miR-148a-3p/WNT1 pathway. J Cell Mol Med. 2020;24(11):6242–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shang A, Wang W, Gu C, Chen C, Zeng B, Yang Y, et al. Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils. J Experimental Clin cancer Research: CR. 2019;38(1):411.

    Article  PubMed Central  Google Scholar 

  30. Feng H, Zhao F, Luo J, Xu S, Liang Z, Xu W, et al. Long non-coding RNA HOTTIP exerts an oncogenic function by regulating HOXA13 in nasopharyngeal carcinoma. Mol Biol Rep. 2023;50(8):6807–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Abdi E, Latifi-Navid S, Zahri S, Kholghi-Oskooei V, Mostafaiy B, Yazdanbod A, et al. SNP-SNP interactions of oncogenic long non-coding RNAs HOTAIR and HOTTIP on gastric cancer susceptibility. Sci Rep. 2020;10(1):16763.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ali MA, Shaker OG, Ezzat EM, Gaber SN, Hassan EA, Abdelwahed MY, et al. Association between rs1859168/HOTTIP expression level and colorectal Cancer and adenomatous polyposis risk in egyptians. J Interferon Cytokine Res. 2020;40(6):279–91.

    Article  CAS  PubMed  Google Scholar 

  33. Duan F, Jiang J, Song C, Wang P, Ye H, Dai L, et al. Functional long non-coding RNAs associated with gastric cancer susceptibility and evaluation of the epidemiological efficacy in a central Chinese population. Gene. 2018;646(null):227–33.

    Article  CAS  PubMed  Google Scholar 

  34. Hu P, Qiao O, Wang J, Li J, Jin H, Li Z, et al. 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 

  35. Yan X, Jia H, Zhao J. LncRNA MEG3 attenuates the malignancy of retinoblastoma cells through inactivating PI3K /Akt/mTOR signaling pathway. Exp Eye Res. 2023;226(null):109340.

    Article  Google Scholar 

  36. Lv D, Bi Q, Li Y, Deng J, Wu N, Hao S, et al. Long non–coding RNA MEG3 inhibits cell migration and invasion of non–small cell lung cancer cells by regulating the miR–21–5p/PTEN axis. Mol Med Rep. 2021;23(3):null.

    Article  Google Scholar 

  37. Tao P, Yang B, Zhang H, Sun L, Wang Y, Zheng W. The overexpression of lncRNA MEG3 inhibits cell viability and invasion and promotes apoptosis in ovarian cancer by sponging miR-205-5p. Int J Clin Exp Pathol. 2020;13(5):869–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Shan G, Tang T, Xia Y, Qian HJ. MEG3 interacted with miR-494 to repress bladder cancer progression through targeting PTEN. J Cell Physiol. 2020;235(2):1120–8.

    Article  CAS  PubMed  Google Scholar 

  39. Zhou C, Cao H, Meng X, Zhang Q. Lnc-MEG3 inhibits invasion, migration, and epithelial- mesenchymal transition of nasopharyngeal carcinoma cells by regulating sequestosome 1. Head and neck-journal for the sciences and specialties of the head and neck. 2022;44(1):201–11.

  40. Gao X, Li X, Zhang S, Wang X. The Association of MEG3 Gene rs7158663 Polymorphism With Cancer Susceptibility. Frontiers in oncology. 2021;11(null):796774.

  41. Kong X, Yang S, Liu C, Tang H, Chen Y, Zhang X, et al. Relationship between MEG3 gene polymorphism and risk of gastric cancer in Chinese population with high incidence of gastric cancer. Biosci Rep. 2020;40(11):null.

    Article  Google Scholar 

  42. Shaker O, Ayeldeen G, Abdelhamid A. The Impact of Single Nucleotide Polymorphism in the Long Non-coding MEG3 Gene on MicroRNA-182 and MicroRNA-29 Expression Levels in the Development of Breast Cancer in Egyptian Women. Frontiers in genetics. 2021;12(null):683809.

  43. Zhuo ZJ, Zhang R, Zhang J, Zhu J, Yang T, Zou Y, et al. Associations between lncRNA MEG3 polymorphisms and neuroblastoma risk in Chinese children. Aging. 2018;10(3):481–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yang Z, Li H, Li J, Lv X, Gao M, Bi Y, et al. Association between Long Noncoding RNA MEG3 polymorphisms and Lung Cancer susceptibility in Chinese Northeast Population. DNA Cell Biol. 2018;37(10):812–20.

    Article  CAS  PubMed  Google Scholar 

  45. Hu JC, Wang SS, Chou YE, Chiu KY, Li JR, Chen CS, et al. Associations between LncRNA MALAT1 polymorphisms and lymph node metastasis in prostate Cancer. Diagnostics (Basel Switzerland). 2021;11(9):null.

    Google Scholar 

  46. Yuan LT, Yang YC, Lee HL, Shih PC, Chen LH, Tang CH, et al. Genetic polymorphisms of lncRNA LINC00673 as predictors of Hepatocellular Carcinoma Progression in an Elderly Population. Int J Mol Sci. 2022;23(21):null.

    Article  Google Scholar 

  47. Hsieh MH, Wu YL, Tsao TC, Huang YW, Lin JC, Lee CY, et al. Impact of LncRNA GAS5 Genetic variants and the epidermal growth factor receptor phenotypes on the clinicopathological characteristics of lung adenocarcinoma patients. Int J Environ Res Public Health. 2022;19(16):null.

    Article  Google Scholar 

  48. Ali MA, Shaker OG, Alazrak M, AbdelHafez MN, Khalefa AA, Hemeda NF, et al. Association analyses of a genetic variant in long non-coding RNA MEG3 with breast cancer susceptibility and serum MEG3 expression level in the Egyptian population. Cancer Biomarkers. 2020;28(1):49–63.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by Guangxi Zhuang Autonomous Region Health Commission project (Grant Nos. Z20201011).

Author information

Authors and Affiliations

Authors

Contributions

XL, YW and RH organized and designed the study. DL, YH and HL collected samples. XL, YH and HL contributed to DNA and RNA extraction. XL, YW and RH contributed to analysis and interpretation of data. XL and YW wrote the manuscript. XL and RH revised the manuscript. All authors reviewed/finalized the manuscript and approved the submission of the article.

Corresponding author

Correspondence to Xiaoxia Lao.

Ethics declarations

Ethics approval and consent to participate

The studies involving human participants were reviewed and approved by committee of Minzu Hospital of Guangxi Zhuang Autonomous Region. The patients/participants provided their written informed consent to participate in this study.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

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

Lao, X., Wang, Y., Huang, R. et al. Genetic variants of LncRNAs HOTTIP and MEG3 influence nasopharyngeal carcinoma susceptibility and clinicopathologic characteristics in the Southern Chinese population. Infect Agents Cancer 19, 32 (2024). https://doi.org/10.1186/s13027-024-00591-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13027-024-00591-6

Keywords