Skip to main content
Journal of Dental Sciences logoLink to Journal of Dental Sciences
. 2024 May 26;19(4):2074–2081. doi: 10.1016/j.jds.2024.05.001

The association between the expression level of nuclear paraspeckle assembly transcript 1 and the survival rate of head and neck cancer patients after treatment

Nan-Chin Lin a,b,c,, Shih-Min Hsia d,, Thanh-Hien Vu Nguyen a, Tong-Hong Wang e, Kuo-Ting Sun a, Kuo-Chou Chiu f,g, Yin-Hwa Shih h,⁎⁎, Tzong-Ming Shieh a,
PMCID: PMC11437243  PMID: 39347098

Abstract

Background/purpose

The long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) exhibits diverse and complicated functions in cancer progression. Despite reports suggesting both tumor-suppressive and oncogenic effects in various cancers, its specific role in head and neck squamous cell carcinoma (HNSCC) remains unclear. This study aimed to investigate the association between NEAT1 expression levels and survival outcomes in HNSCC patients.

Materials and methods

Paired tissue samples of tumor and non-cancerous matching tissues (NCMT) from 92 HNSCC patients were collected. NEAT1 expression was analyzed using RT-qPCR. Clinical characteristics, treatment received, and survival rates of the patients were assessed to determine the correlation with NEAT1 expression and explore its association with alcohol, betel quid, and cigarette use. Additionally, we examined the effect of arecoline on NEAT1 expression in normal human oral keratinocytes (NHOK) and fibroblasts (NHOF).

Results

The study revealed a significant downregulation of NEAT1 expression in oral cancer tissues compared to NCMT. Meanwhile, arecoline increased NEAT1 expression in NHOK and NHOF cells. However, patients with downregulated NEAT1 expression exhibited higher overall survival rates, particularly in those who did not receive chemotherapy or radiotherapy.

Conclusion

NEAT1 expression levels are associated with survival outcomes in HNSCC patients, with upregulated expression indicating a worse prognosis, suggesting this lncRNA might contribute to cancer aggressiveness, especially in the absence of active treatment. These findings indicate NEAT1 may serve as a potential prognostic biomarker in HNSCC, but further research is required to elucidate its role in cancer progression and its potential as a therapeutic target.

Keywords: Nuclear paraspeckle assembly transcript 1, Long non-coding RNA, Head and neck squamous cell carcinoma, Survival outcomes, Oral cancer

Introduction

Head and neck squamous cell carcinoma (HNSCC) poses a significant public health threat, claiming numerous lives globally. HNSCC refers to a broad category of malignancies originating from the squamous cells lining the mucosal surfaces of the head and neck region, including the oral cavity, pharynx, and larynx, encompassing tumors affecting the upper aerodigestive tract. With its aggressive nature and high mortality rates, roughly 890,000 new cases of HNSCC have been reported annually, making HNSCC the sixth most frequent cancer in the world and accounting for 450,000 deaths each year.1, 2, 3, 4 Therefore, understanding the intricate mechanisms underlying its development and progression is crucial for improving patient outcomes. Unraveling the intricate mechanisms fueling its development and progression holds the key to improving patient outcomes, and within this pursuit, the realm of long non-coding RNA (lncRNA) offers promising avenues for exploration.5

Certain lncRNAs have been found to prevent tumor growth, whereas other lncRNAs act as oncogenes, promoting the formation and metastasis of cancer.6 Among these, lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) has emerged as a fascinating player, showcasing oncogenic activity in various cancers.7 Its association with oncogenic activity across various cancers, including breast, liver, thyroid, prostate, and pancreas, has ignited substantial research interest.8, 9, 10, 11, 12 Our prior study using microarray analysis revealed that overexpression of NEAT1 is associated with poor survival rate in oral squamous cell carcinoma (OSCC).13 Additionally, other previous study reported that NEAT1 is involved in the carcinogenic development of laryngeal squamous cell carcinoma (LSCC) and contributes to the disease's aggressiveness.14 However, the landscape of NEAT1's involvement in HNSCC on a general scale still remains unknown.

We investigated the expression patterns of NEAT1 in cancer tissues compared to their healthy counterparts in Taiwanese HNSCC patients. Moreover, we examined the associations between NEAT1 levels and clinicopathological features, revealing its potential impact on tumor progression and patient prognosis. The intricate nature of NEAT1's involvement in HNSCC calls for more research to fully understand its complicated role in various head and neck cancer subtypes and cell lines under diverse physiological conditions.

Materials and methods

Subjects

92 pairs of non-cancerous matched tissues (NCMT) and tissues from individuals with HNSCC were provided from the tissue bank at Changhua Christian Hospital. The study received ethical approval from the Institutional Review Board (Approval number: 200501). Tissue samples were collected and immediately frozen in liquid nitrogen for further examination. A thorough selection method was used on frozen sections to guarantee that HNSCC samples contained more than 70% tumor cells, as required for the study.13 Table 1 provides comprehensive clinicopathological data, including age, gender, lesion location, differentiation, tumor, node, metastasis (TNM) stage, pathological stage, treatment methods, and survival status. Additionally, alcohol consumption, betel quid, and cigarette usage in connection with NEAT1 RNA expression in HNSCC patients are included in Table 3.

Table 1.

Subjects and clinical characteristics of HNSCC.

HNSCC (n = 92, mean of age = 57.03 ± 10.33)
Gender
Male 88
Female 4
Type
Oral cancer 35
Oropharynx 22
Laryngopharynx 6
Larynx 29
Differentiation
No record 5
Well 9
Moderately 74
Poorly 4
TNM
No record 16
T0 0
T1 22
T2 21
T3 7
T4 26
TNM
No record 18
N = 0 34
N > 0 40
Stage
No record 16
Stage I 15
Stage II 5
Stage III 11
Stage IV 45
Chemotherapy
No 31
Yes 61
Radiotherapy
No 36
Yes 56
Survival status
Live 36
Die 56

HNSCC, head and neck squamous cell carcinoma.

TNM, tumor, node, metastasis stage.

Table 3.

Effect of alcohol, betel quid, and cigarette on NEAT1 RNA expression in HNSCC and OSCC.

Down (<1.2) Up ( ≥ 1.2) P OR
HNSCC (n = 92)
Alcohol
No 22 13 0.6374 1.231
Yes 33 24
Betel quid
No 22 16 0.7567 0.8750
Yes 33 21
Cigarette
No 16 9 0.6143 1.276
Yes
39
28


OSCC (n = 35)
Alcohol
No 11 5 0.4917 0.5867
Yes 15 4
Betel quid
No 11 3 0.6357 1.467
Yes 15 6
Cigarette
No 8 3 0.8864 0.8889
Yes 18 6

HNSCC, head and neck squamous cell carcinoma.

TNM, tumor, node, metastasis stage.

OSCC, oral squamous cell carcinoma.

OR, odds ratio.

The relative NEAT1 RNA expression median value of 1.2-fold was used as the cut-off value to determine the up- or down-regulation expression group.

Down, down-regulation.

Up, up-regulation.

Cell culture

Normal human oral keratinocytes (NHOK) and normal human oral fibroblasts (NHOF) were cultured in keratinocyte serum-free medium (Gibco BRL, Gaithersburg, MD, USA) and Dulbecco's Modified Eagle Medium (DMEM) (Gibco BRL) plus 10% fetal bovine serum (FBS), respectively, following the earlier protocols.15 The cell lines were cultured in an incubator at 37 °C and 5% CO2. To investigate the effects of arecoline, a major alkaloid presents in betel quid, on the expression of NEAT1 in the NHOK and NHOF cells, we used different doses of arecoline (50 and 100 μg/ml) to treat the cells for 24 h before harvesting and extracting their RNA.

RNA extraction from tissues and cells

Total RNA was extracted and purified using a TRI Reagent RNA isolation kit (Molecular Research Center, Cincinnati, OH, USA), with the process adjusted according to the manufacturer's instructions. The RNA concentration was determined by using NanoVue Plus spectrophotometer (General Electric Company, Boston, MA, USA), and running electrophoresis in 1% agarose gel to confirm the RNA purity. RNA samples were later processed with DNase I (Stratagene, La Jolla, CA, USA) to eliminate contaminant DNA. Subsequently, two micrograms of total RNA were converted into complementary DNA (cDNA) by using reverse transcriptase and a random primer.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

To analyze NEAT1 RNA expression, quantitative PCR (qPCR) was performed by a StepOnePlus™ Real-Time PCR System (Applied Biosystems, Waltham, MA, USA). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal control. The primer sequences utilized in this investigation are shown below:

  • NEAT1: Forward: 5’-CTTCCTCCCTTTAACTTATCCATTCAC-3’,
    • Reverse: 5’-CTCTTCCTCCACCATTACCAACAATAC-3’;
  • GAPDH: Forward: 5’-TGGTATCGTGGAAGGACTCATGAC-3’,
    • Reverse: 5’-ATGCCAGTGAGCTTCCCGTTCAGC-3’.

These forward and reverse primer sequences were designed based on the sequences used in the previously published study.16,17 The PowerUp SYBR Green Master Mix (Applied Biosystems) was used for PCR, and genes were amplified using 40 cycles of 95 °C for 15 s and 60 °C for 1 min. To ensure analysis repeatability, triplicate PCRs were performed for each sample. Samples with conflicting findings were removed from the final analysis. NEAT1 RNA expression in the NCMT and HNSCC samples was measured by -ΔCt (CtGAPDH–CtNEAT1), the -ΔCt value represents the relative expression level of NEAT1 normalized to GAPDH. The median value of 1.2-fold for relative RNA expression of NEAT1 between HNSCC and NCMT was used as a cut-off value to determine if one subject belonged to the NEAT1 up- or down-regulation expression group.

Statistical analysis

Data were analyzed using GraphPad Prism (v9, GraphPad Software Inc., La Jolla, CA, USA) to calculate odds ratios, Fisher's exact test, unpaired t-test, Mann–Whitney U test, and logistic regression. Survival analysis was carried out utilizing Kaplan–Meier survival curves, with significance determined by the log-rank test. Differences were considered statistically significant at a P-value <0.05.

Results

Demographic and clinical characteristics of HNSCC patients

A total of 92 patients with HNSCC were included in this study, with an average age of 57.03 ± 10.33 years. The majority are male (88); only four patients are female. Most patients had oral cancer (35), followed by oropharyngeal cancer (22), and then laryngeal cancer (29) and laryngopharyngeal cancer (6). Detailed demographic and clinical information was summarized in Table 1.

NEAT1 RNA expression in HNSCC subtypes and NEAT1 RNA was down-regulated in OSCC

The comparison of NEAT1 RNA expression levels between non-cancerous matched tissue (NCMT) and HNSCC tissue, as depicted in Fig. 1A, revealed no statistically significant differences. Based on the provided data in Table 2, most oral cancer cases (26 of 35) have NEAT1 RNA expression being decreased (expression fold change less than 1.2 compared to NCMT). While significant downregulation of NEAT1 RNA was observed in oral cancer, the remaining 67 cases of other HNSCC (oropharyngeal, laryngopharyngeal, and laryngeal cancers) did not exhibit this trend (Fig. 1B). The distribution of “down” (29 cases) and “up” (28 cases) groups remained statistically comparable in these other HNSCC subtypes (Table 2).

Figure 1.

Figure 1

Expression levels of NEAT1 RNA in HNSCC. A. Comparison of NEAT1 RNA expression levels between non-cancerous matched tissue (NCMT) and HNSCC tissue (P = 0.5107). Mann–Whitney U test was used to analyze the data. B.NEAT1 RNA expression levels across different HNSCC subtypes.

Table 2.

NEAT1 RNA expression in HNSCC.

HNSCC (n = 92) Cut off at the median of 1.2-fold
Down (<1.2) Up (≥1.2) P OR
Age(mean = 57)
<57 30 16 0.2877 1.575
≥57 25 21
Type
Oral cancer 26 9 0.0262 2,789
Oropharynx 11 11
Laryngopharynx 2 4
Larynx 16 13
Differentiation
No record 4 1
Well 6 3 0.6047 1,467
Moderately 42 32
Poorly 3 1
TNM, T
No record 11 5
T0+T1+T2 23 20 0.3745 0.6571
T3+T4 21 12
TNM, N
No record 10 8
N = 0 23 11 0.2667 1.711
N > 0 22 18
Stage
No record 11 5
Stage I + II + III 18 13 0.9801 1.012
Stage IV 26 19

HNSCC, head head and neck squamous cell carcinoma.

TNM, tumor, node, metastasis stage.

OR, odds ratio.

The relative NEAT1 RNA expression median value of 1.2-fold was used as the cut-off value to determine the up- or down-regulation expression group

Down, down-regulation.

Up, up-regulation.

Fisher's exact test was used to analyze the data.

NEAT1 RNA expression was not significantly associated with alcohol, cigarette use, or betel quid chewing in HNSCC patients but was upregulated in NHOK and NHOF cells under arecoline treatment in vitro

The expression of NEAT1 RNA is downregulated in the oral cancer subgroup, contrary to other head and neck cancer sites. Therefore, the association between risk factors for oral cancer such as alcohol consumption, betel quid chewing, cigarette use habits, and NEAT1 RNA expression was compared. Table 3 includes data on alcohol consumption, betel quid, and cigarette usage in HNSCC patients with NEAT1 RNA expression levels. The data shows no significant association between those lifestyle habits and NEAT1 RNA expression. Conversely, Fig. 2 illustrates arecoline treatment produced NEAT1 RNA upregulation in NHOK and NHOF cells. While arecoline treatment at both 50 and 100 μg/ml caused a significant increase in NEAT1 RNA expression in the NHOK cell line (Fig. 2A), only the 100 μg/ml dose can induce significant upregulation of NEAT1 RNA expression in NHOF (Fig. 2B).

Figure 2.

Figure 2

Arecoline treatment induced NEAT1 RNA upregulation in normal cells. A. Arecoline treatment caused a significant increase in NEAT1 RNA expression in normal human oral keratinocytes (NHOK). B. Arecoline treatment at 100 μg/ml significantly increased NEAT1 RNA expression in normal human oral fibroblasts (NHOF). NEAT1 RNA expression levels in NHOK and NHOF cells were determined by RT-qPCR at 24 h after treatment. All experiments were performed in triplicate. Data are expressed as mean ± SD. ∗P < 0.05, ∗∗P < 0.01. Unpaired t-test was used to analyze the data.

Upregulation of NEAT1 is associated with reduced survival probability in HNSCC patients

Table 4 demonstrates the association between NEAT1 RNA expression, treatment received, and survival status in HNSCC patients. Overall, patients with down-regulated NEAT1 RNA expression had greater survival rates, regardless of treatment received. Significant higher survival with down-regulation of NEAT1 is also observed in patients who did not receive chemotherapy (P = 0.0468). Similar pattern, but the difference in survival rate between the NEAT1 RNA down- and up-regulation groups is more pronounced in patients who did not receive radiotherapy (P = 0.0055). In addition, this trend can be seen in chemotherapy-received patients but has no statistical significance (P = 0.1342), while no difference was found in the survival rate in the radiotherapy group.

Table 4.

Association between NEAT1 RNA expression and treatment with survival probability in HNSCC.

HNSCC (n = 92) Cut off at the fold median of 1.2-fold
Down (<1.2) Up (≥1.2) P OR
Survival status
Live 27 9 0.0170 3.000
Die 28 28
No radiotherapy treatment 23 14 0.7026 1.181
Live 14 2 0.0055 9.333
Die 9 12
Radiotherapy treatment 32 23
Live 13 7 0.4384 1.564
Die 19 16
No chemotherapy treatment 20 11 0.5092 1.351
Live 11 2 0.0468 5.500
Die 9 9
Chemotherapy treatment 35 26
Live 16 7 0.1342 2.286
Die 19 19

HNSCC, head and neck squamous cell carcinoma.

TNM, tumor, node, metastasis stage.

OR, odds ratio.

The relative NEAT1 RNA expression median value of 1.2-fold was used as the cut-off value to determine the up- or down-regulation expression group.

Down, down-regulation.

Up, up-regulation.

Fisher's exact test was used to analyze the data.

In agreement with data from Table 4, the Kaplan–Meier plots in Fig. 3 illustrate the survival probability of HNSCC patients in relation to NEAT1 RNA expression. A reduced survival probability was observed in the HNSCC patients with upregulated NEAT1 RNA expression (Fig. 3A). On one hand, there was no significant difference in survival probability among up- and down-regulated NEAT1 RNA expression groups in the patients who received chemotherapy and radiotherapy treatment (Fig. 3B–D). On the other hand, non-receiving chemotherapy and/or radiotherapy treatment patients with a lower survival probability expressed considerably higher levels of the NEAT1 gene (Fig. 3C–E). That indicates higher levels of NEAT1 RNA expression were associated with a worse survival probability, especially in HNSCC patients who did not get chemotherapy and radiotherapy.

Figure 3.

Figure 3

Kaplan–Meier plot of the survival probability of HNSCC patients in relation to NEAT1 RNA expression. The red curve represents the up-regulation group, and the blue curve is the down-regulation group of NEAT1 RNA expression. A reduced survival probability was observed in the HNSCC patients with increased NEAT1 RNA expression (A). There was no significant difference in survival probability between “up” and “down” NEAT1 RNA expression groups in the patients who received chemotherapy and radiotherapy treatment (B, D). In the non-chemotherapy and non-radiotherapy patients, the up-regulated NEAT1 group showed a significantly lower survival probability compared to the down-regulated group (C, E). n, sample size. ∗P < 0.05, ∗∗P < 0.01. Kaplan–Meier survival curves and the log-rank test were used to analyze the data.

Discussion

Since its discovery, NEAT1 has been linked to upregulation in a variety of cancers. However, there are contradictions since NEAT1 may also act as a tumor suppressor in several cancers, including breast invasive carcinoma, esophageal carcinoma, nasopharyngeal carcinoma, and acute myeloid leukemia.18, 19, 20, 21 This present study found downregulation of NEAT1 expression compared to normal tissues in most cases of oral cancer. Meanwhile, we did not observe significant alterations in NEAT1 expression in other HNSCC subtypes, including oropharyngeal, laryngopharyngeal, and laryngeal cancers, possibly due to the small sample size of these cancers. Nonetheless, this discrepancy demonstrates the potential heterogeneity within the HNSCC spectrum, emphasizing the importance of conducting subgroup analyses in future studies with larger sample sizes. The observed downregulation of NEAT1 in oral cancer, while not evident in other subtypes, prompts exploration into potential variations in tumorigenesis pathways or unique genetic alterations specific to oral cancer, demanding further investigation in subsequent investigations.

Moreover, our findings suggest that NEAT1 expression holds promise as a potential prognostic biomarker in HNSCC. Patients exhibiting upregulated NEAT1 demonstrated poor overall survival rates, a trend that is substantially prominent in non-chemo-radiotherapy-received groups. The intriguing aspect of our study lies in the correlation between the upregulation of NEAT1 RNA expression and reduced survival probability in HNSCC patients. As mentioned earlier, the downregulation of NEAT1 was found in most cases of oral cancer, but it was contrarily associated with improving survival outcomes in overall HNSCC cases. Thus, our findings indicate that the upregulation of NEAT1 RNA expression may serve as an unfavorable marker in HNSCC, indicating a potential link between elevated NEAT1 levels and reduced survival probability. However, to establish the clinical utility of NEAT1 as a prognostic marker, further validation through larger cohorts and prospective studies is imperative.

NEAT1 is an important lncRNA involved in various nuclear functions, particularly within paraspeckles. It influences mRNA processing, gene regulation, and contributes to diverse cellular processes.22 The complex role of NEAT1 in HNSCC survival remains unclear, necessitating exploration of its interactions with other genes or pathways involved in cancer progression. In nasopharyngeal carcinoma, NEAT1 knockdown has been reported to be able to inhibit cell proliferation and migration by impeding Wnt/β-catenin signaling. The study also found that NEAT1 knockdown inhibited Wnt/β-catenin signaling via miR-34a-5p. NEAT1 binds directly to miR-34a-5p, diminishing its expression. In addition, NEAT1 knockdown lowered cell migration, invasion, proliferation, and epithelial-to-mesenchymal transition (EMT) via miR-34a-5p.23 Moreover, NEAT1 upregulation may activate miR-124/NF-κB signaling modulation, hinting at a potential role in the tumorigenesis and progression pathways in HNSCC.24 Conversely, downregulation of NEAT1 could impede this activation, potentially contributing to improved survival outcomes. Additionally, high expression levels of the lncRNA NEAT1 were associated with unfavorable overall survival outcomes in breast cancer patients, while inhibition of NEAT1 expression led to the suppression of the EMT process via increased levels of miR-146 b-5p.25 The downregulation of NEAT1 could potentially disrupt this interaction, acting as a hindrance to cancer cell proliferation. A study in LSCC revealed that NEAT1 functions as a sponge for miR-107, resulting in enhancing the expression of cyclin-dependent kinase 6 (CDK6), a key member of the CDK family strongly associated with the progression of head and neck squamous cell carcinoma.14,26 In lung cancer, NEAT1 has been associated with facilitating cyclin D1 expression, thereby driving cell cycle progression.27 Consequently, downregulation of NEAT1 may disturb the cell cycle of tumor growth, presenting a potential target for therapeutic intervention in HNSCC. Moreover, NEAT1's involvement in epithelial–mesenchymal transition, a critical process for cancer invasion and metastasis, has been documented. Downregulation of NEAT1 might hinder EMT, restricting cancer spread within the head and neck cancers context.28,29 NEAT1 has been linked to the promotion of matrix metalloproteinase (MMP) expression, enzymes involved in extracellular matrix degradation, and metastasis. Silencing of NEAT1 could potentially reduce MMP activity, thereby limiting the invasive potential of cancer.30,31 Some studies suggested that NEAT1 might play a role in suppressing the anti-tumor immune response via increasing CD8+ T cell apoptosis, which contributes to the restraint of T-cell activity in tumor elimination. Repression of NEAT1 in HNSCC could potentially enhance the immune system's activity against cancer cells.32 These multifaceted roles of NEAT1 in diverse molecular pathways highlight its intricate involvement in the complex landscape of cancer progression. Hence, unraveling the underlying mechanisms related to NEAT1 expression and its impact on survival outcomes in HNSCC is critical for achieving a thorough comprehension and suitable therapeutic strategy for these malignancies.

In other aspects, we found that there was no statistical significance between the use of cigarettes, alcohol, and betel quid and the expression of NEAT1 in patients with head and neck cancer. However, in addition to the observed downregulation of NEAT1 expression in oral cancer, our investigation also revealed arecoline, a major alkaloid in betel quid, could induce NEAT1 expression in NHOK and NHOF. In other words, NEAT1 expression exhibited an elevation in reaction against the exposure of arecoline, a well-known carcinogen, thus suggesting a potential protective mechanism against oral carcinogenesis. Hence, it is plausible that NEAT1 may act as a tumor suppressor in oral cancer, as earlier mentioned NEAT1's involvement in various tumor-suppressive pathways in other cancer types.32 Therefore, further investigation is needed to identify under what circumstances or factors modulate NEAT1's activities as a tumor oncogene or suppressor.

In conclusion, this study provides evidence for the potential prognostic significance of NEAT1 expression in HNSCC. The observed downregulation of NEAT1 associated with improved survival outcomes in HNSCC suggests that NEAT1 may serve as a valuable biomarker for classification, prognosis, and treatment approach. However, further research, including larger cohort studies and investigations into the underlying mechanisms, is essential to validate the clinical utility of NEAT1 and explore its potential as a therapeutic target in distinct cases of HNSCC.

Declaration of competing interest

The authors have no conflicts of interest relevant to this article.

Acknowledgments

Experiments and data analysis were performed in part through the use of the Medical Research Core Facilities Center, Office of Research & Development at China Medical University, Taichung, Taiwan, R.O.C.). This study was funded by the China Medical University, grant numbers CMU112-MF-41 and CMU112-S-51, and the Ministry of Science and Technology (MOST), grant numbers MOST 111-2314-B-039-027-MY3.

Contributor Information

Yin-Hwa Shih, Email: evashih@asia.edu.tw.

Tzong-Ming Shieh, Email: tmshieh@mail.cmu.edu.tw.

References

  • 1.Johnson D.E., Burtness B., Leemans C.R., Lui V.W.Y., Bauman J.E., Grandis J.R. Head and neck squamous cell carcinoma. Nat Rev Dis Prim. 2020;6:92. doi: 10.1038/s41572-020-00224-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
  • 3.Ferlay J., Colombet M., Soerjomataram I., et al. Estimating the global cancer incidence and mortality in 2018: globocan sources and methods. Int J Cancer. 2019;144:1941–1953. doi: 10.1002/ijc.31937. [DOI] [PubMed] [Google Scholar]
  • 4.Chan C.Y., Lien C.H., Lee M.F., Huang C.Y. Quercetin suppresses cellular migration and invasion in human head and neck squamous cell carcinoma (HNSCC) Biomedicine. 2016;6:15. doi: 10.7603/s40681-016-0015-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Grixti J.M., Ayers D. Long noncoding RNAs and their link to cancer. Noncoding RNA Res. 2020;5:77–82. doi: 10.1016/j.ncrna.2020.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bhan A., Soleimani M., Mandal S.S. Long noncoding RNA and cancer: a new paradigm. Cancer Res. 2017;77:3965–3981. doi: 10.1158/0008-5472.CAN-16-2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hussain M.S., Gupta G., Afzal M., et al. Exploring the role of lncRNA NEAT1 knockdown in regulating apoptosis across multiple cancer types: a review. Pathol Res Pract. 2023;252 doi: 10.1016/j.prp.2023.154908. [DOI] [PubMed] [Google Scholar]
  • 8.Chakravarty D., Sboner A., Nair S.S., et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat Commun. 2014;5:5383. doi: 10.1038/ncomms6383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li X., Wang S., Li Z., et al. The lncRNA NEAT1 facilitates cell growth and invasion via the miR-211/HMGA2 axis in breast cancer. Int J Biol Macromol. 2017;105:346–353. doi: 10.1016/j.ijbiomac.2017.07.053. [DOI] [PubMed] [Google Scholar]
  • 10.Wang Z., Zou Q., Song M., Chen J. NEAT1 promotes cell proliferation and invasion in hepatocellular carcinoma by negative regulating miR-613 expression. Biomed Pharmacother. 2017;94:612–618. doi: 10.1016/j.biopha.2017.07.111. [DOI] [PubMed] [Google Scholar]
  • 11.Huang B., Liu C., Wu Q., et al. Long non-coding RNA NEAT1 facilitates pancreatic cancer progression through negative modulation of miR-506-3p. Biochem Biophys Res Commun. 2017;482:828–834. doi: 10.1016/j.bbrc.2016.11.120. [DOI] [PubMed] [Google Scholar]
  • 12.Li J.H., Zhang S.Q., Qiu X.G., Zhang S.J., Zheng S.H., Zhang D.H. Long non-coding RNA NEAT1 promotes malignant progression of thyroid carcinoma by regulating miRNA-214. Int J Oncol. 2017;50:708–716. doi: 10.3892/ijo.2016.3803. [DOI] [PubMed] [Google Scholar]
  • 13.Lin N.C., Hsia S.M., Wang T.H., et al. The relation between NEAT1 expression level and survival rate in patients with oral squamous cell carcinoma. J Dent Sci. 2022;17:361–367. doi: 10.1016/j.jds.2021.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wang P., Wu T., Zhou H., et al. Long noncoding RNA NEAT1 promotes laryngeal squamous cell cancer through regulating miR-107/CDK6 pathway. J Exp Clin Cancer Res. 2016;35:22. doi: 10.1186/s13046-016-0297-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shieh T.M., Lin S.C., Liu C.J., Chang S.S., Ku T.H., Chang K.W. Association of expression aberrances and genetic polymorphisms of lysyl oxidase with areca-associated oral tumorigenesis. Clin Cancer Res. 2007;13:4378–4385. doi: 10.1158/1078-0432.CCR-06-2685. [DOI] [PubMed] [Google Scholar]
  • 16.Yang X., Xiao Z., Du X., Huang L., Du G. Silencing of the long non-coding RNA NEAT1 suppresses glioma stem-like properties through modulation of the miR-107/CDK6 pathway. Oncol Rep. 2017;37:555–562. doi: 10.3892/or.2016.5266. [DOI] [PubMed] [Google Scholar]
  • 17.Kano M., Shimada Y., Kaganoi J., et al. Detection of lymph node metastasis of oesophageal cancer by RT-nested PCR for SCC antigen gene mRNA. Br J Cancer. 2000;82:429–435. doi: 10.1054/bjoc.1999.0938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rheinbay E., Parasuraman P., Grimsby J., et al. Recurrent and functional regulatory mutations in breast cancer. Nature. 2017;547:55–60. doi: 10.1038/nature22992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Duan M.Y., Li M., Tian H., Tang G., Yang Y.C., Peng N.C. Down-regulation of lncRNA NEAT1 regulated by miR-194-5p/DNMT3A facilitates acute myeloid leukemia. Blood Cells Mol Dis. 2020;82 doi: 10.1016/j.bcmd.2020.102417. [DOI] [PubMed] [Google Scholar]
  • 20.Wang Y., Wang C., Chen C., et al. Long non-coding RNA NEAT1 regulates epithelial membrane protein 2 expression to repress nasopharyngeal carcinoma migration and irradiation-resistance through miR-101-3p as a competing endogenous RNA mechanism. Oncotarget. 2017;8:70156–70171. doi: 10.18632/oncotarget.19596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li S., Li J., Chen C., Zhang R., Wang K. Pan-cancer analysis of long non-coding RNA NEAT1 in various cancers. Genes Dis. 2018;5:27–35. doi: 10.1016/j.gendis.2017.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Klec C., Prinz F., Pichler M. Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol Oncol. 2019;13:46–60. doi: 10.1002/1878-0261.12404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ji Y., Wang M., Li X., Cui F. The long noncoding RNA NEAT1 targets miR-34a-5p and drives nasopharyngeal carcinoma progression via Wnt/β-catenin signaling. Yonsei Med J. 2019;60:336–345. doi: 10.3349/ymj.2019.60.4.336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cheng N., Guo Y. Long noncoding RNA NEAT1 promotes nasopharyngeal carcinoma progression through regulation of miR-124/nf-κb pathway. OncoTargets Ther. 2017;10:5843–5853. doi: 10.2147/OTT.S151800. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 25.Li S., Hao J., Hong Y., Mai J., Huang W. Long non-coding RNA NEAT1 promotes the proliferation, migration, and metastasis of human breast-cancer cells by inhibiting miR-146b-5p expression. Cancer Manag Res. 2020;12:6091–6101. doi: 10.2147/CMAR.S252295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Poomsawat S., Sanguansin S., Punyasingh J., Vejchapipat P., Punyarit P. Expression of cdk6 in head and neck squamous cell carcinoma. Clin Oral Invest. 2016;20:57–63. doi: 10.1007/s00784-015-1482-8. [DOI] [PubMed] [Google Scholar]
  • 27.Sun C., Li S., Zhang F., et al. Long non-coding RNA NEAT1 promotes non-small cell lung cancer progression through regulation of miR-377-3p-e2f3 pathway. Oncotarget. 2016;7:51784–51814. doi: 10.18632/oncotarget.10108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen Y., Li J., Xiao J.K., Xiao L., Xu B.W., Li C. The lncRNA NEAT1 promotes the epithelial-mesenchymal transition and metastasis of osteosarcoma cells by sponging miR-483 to upregulate STAT3 expression. Cancer Cell Int. 2021;21:90. doi: 10.1186/s12935-021-01780-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lu Y., Li T., Wei G., et al. The long non-coding RNA NEAT1 regulates epithelial to mesenchymal transition and radioresistance in through miR-204/ZEB1 axis in nasopharyngeal carcinoma. Tumour Biol. 2016;37:11733–11741. doi: 10.1007/s13277-015-4773-4. [DOI] [PubMed] [Google Scholar]
  • 30.Gong Z., Shen G., Huang C., Zhang J., Ji J. Downregulation of lncRNA NEAT1 inhibits the proliferation of human cutaneous squamous cell carcinoma in vivo and in vitro. Ann Transl Med. 2022;10:79. doi: 10.21037/atm-21-6916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Xiao P., Zhu X., Sun J., et al. Lncrna NEAT1 regulates chondrocyte proliferation and apoptosis via targeting miR-543/PLA2G4A axis. Hum Cell. 2021;34:60–75. doi: 10.1007/s13577-020-00433-8. [DOI] [PubMed] [Google Scholar]
  • 32.Yan K., Fu Y., Zhu N., et al. Repression of lncRNA NEAT1 enhances the antitumor activity of CD8+T cells against hepatocellular carcinoma via regulating miR-155/Tim-3. Int J Biochem Cell Biol. 2019;110:1–8. doi: 10.1016/j.biocel.2019.01.019. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Dental Sciences are provided here courtesy of Association for Dental Sciences of the Republic of China

RESOURCES