Skip to main content
International Journal of Ophthalmology logoLink to International Journal of Ophthalmology
. 2020 Aug 18;13(8):1195–1201. doi: 10.18240/ijo.2020.08.04

LncRNA SNHG15 predicts poor prognosis in uveal melanoma and its potential pathways

Xue Wu 1,2,3, Xiao-Feng Li 1,2,3, Qian Wu 4, Rui-Qi Ma 1,2,3, Jiang Qian 1,2,3, Rui Zhang 1,2,3
PMCID: PMC7387894  PMID: 32821672

Abstract

AIM

To evaluate the role of long noncoding RNA (lncRNA) SNHG15 and its potential pathways in uveal melanoma (UM).

METHODS

The SNHG15 mRNA expression level and corresponding clinicopathological characteristics of 80 patients with UM were obtained from the Cancer Genome Atlas (TCGA) database and further analyzed. The SPSS 24.0 statistical software package was used for statistical analyses. To investigate the potential function of SNHG15 in UM, we conducted in-depth research on Gene Set Enrichment Analysis (GSEA).

RESULTS

The univariate analysis revealed that the age, tumor diameter, pathological type, extrascleral extension, cancer status, and high expression of SNHG15 were statistical risk factors for death from all causes. The multivariate analysis suggested that the mRNA expression level of SNHG15 was an independent risk factor for death from all causes, as was age and pathological type. Kaplan-Meier survival analysis confirmed that UM patients with high SNHG15 expression might have a poor prognosis. In addition, SNHG15 was significantly differentially expressed in the different groups of tumor pathologic stage, metastasis and living status. Besides, the logistic regression analysis indicated that high SNHG15 expression group in UM was significantly associated with cancer status, pathologic stage, metastasis, and living status. Moreover, the GSEA indicated the potential pathways regulated by SNHG15 in UM.

CONCLUSION

Our research suggests that SNHG15 may play a vital role as a potential marker in UM that predicts poor prognosis. Besides, GSEA indicates the underlying signaling pathways enriched differentially in SNHG15 high expression phenotype.

Keywords: SNHG15, uveal melanoma, the Cancer Genome Atlas, pathology, prognosis, Gene Set Enrichment Analysis

INTRODUCTION

Uveal melanoma (UM), the most common intraocular cancer in adult worldwide[1], is a malignant tumor that originates in melanocytes of the choroid plexus, ciliary body, and iris of the eye. At present, despite definitive radiotherapy or removal of the primary lesion, numerous patients eventually develop metastases and subsequently prognosis is significantly poor[2]. In addition, UM tends to metastasize to liver through hematogenous pathway, a distant site relative to their origins in the eye[3]. There is an incubation period between the enucleation of the primary tumor and the emergence of metastasis, which can range from a few months to several decades[4][5]. Despite the advancement of UM management, there are currently no effective therapy once the metastases occurred[6]. Therefore, close follow-up and further research on the pathogenesis and novel makers exploration of UM are of great significance for accurate diagnosis, appropriate therapy and prognosis prediction.

Long noncoding RNA (lncRNA), is a class of noncoding transcripts with a length of larger than 200 nucleotides[7], which has been involved widely in biological processes of different cancers, including cell cycle, apoptosis, cell differentiation[8][10]. In the development of UM, lncRNA is also reported to play a vital role in cell cycle, cell proliferation, apoptosis, invasion and autophagy[11][13]. For example, silencing of lncRNA PVT1 prevents the development of UM by impairing microRNA-17-3p-dependent MDM2 upregulation[14]. ZNNT1 can suppress the progression of UM by inducing the expression of crucial autophagy gene[15]. The lncRNA RHPN1-AS1 facilitates the tumorigenesis of UM by influencing cell proliferation and migration[13]. However, the study of vital lncRNAs in UM still remains to be explored.

SNHG15, a novel lncRNA, located on chromosome 7p13[16], is identified to play a key role in many types of human tumors, such as osteosarcoma[17], papillary thyroid carcinoma[18], pancreatic ductal adenocarcinoma[19], colorectal carcinoma[20], hepatocellular carcinoma[21][22], prostate cancer[23], and breast cancer[24]. To our knowledge, the potential impact of SNHG15 on the tumorigenesis of UM seems unclear recently. Thus, the purpose of this study was to evaluate the pivotal role of SNHG15 in the progression of UM. In addition, the relationship between SNHG15 expression and clinicopathologic characteristics in UM was preliminarily demonstrated. To explore the underlying mechanisms of the biological pathways involved in UM, we conducted a research on Gene Set Enrichment Analysis (GSEA).

MATERIALS AND METHODS

Ethical Approval

The study protocol was approved by the Ethics Committee of the Eye & ENT Hospital of Fudan University, and all procedures were complied with the principles of the Declaration of Helsinki. All datasets of our present study were downloaded from an open database TCGA, so there was no written informed consent from participants.

RNA-Sequencing Patient Data and Bioinformatics Analysis

The RNA-Seq gene expression level and clinicopathological characteristics, including 80 cases, were obtained from the official website of the Cancer Genome Atlas (TCGA) UM project (https://portal.gdc.cancer.gov/). Patients with UM were classified as two groups, based on the median SNHG15 expression level (cutoff value=7.94 FPKM). Finally, 80 patients with UM were retained and their clinicopathological characteristics were further analyzed, including the detailed information of age, gender, tumor diameter, thickness, pathological type, extrascleral extension, cancer status, pathological stage, metastasis, living status, SNHG15 expression.

Gene Set Enrichment Analysis

GSEA is a common bioanalysis used to interpret and analyze microarray and other similar data, and to speculate related pathways that can significantly enrich regulatory genes[25]. Through TCGA UM project, we obtained the RNA-Seq gene expression level of 80 UM patients. And the analysis was conducted using GSEA v3.0 software. In this study, according to the association with SNHG15 expression, the ordered gene list was generated firstly by GSEA. Subsequently, GSEA was conducted to clarify statistically significant differences between the two groups with high and low SNHG15 expression. A total of 1000 permutations were performed. The SNHG15 expression level was identified as a phenotype label. The related pathways statistically enriched in each phenotype were selected with the nominal P<0.05 and an false discovery rate (FDR) <0.25.

Statistical Analysis

The SPSS 24.0 statistical software package (SPSS, Inc., USA) was used for statistical analyses. Both the univariate and multivariate analyses using Cox analysis were performed to demonstrate independent prognostic biomarkers for UM patients. The survival curve was generated by conducting Kaplan-Meier method. To compare the significant differences in overall survival (OS), the log-rank test was conducted. The plot chart was performed to visualize the difference of SNHG15 expression level for diverse variables through Graphpad. The relationship between the SNHG15 expression and clinicopathological characteristics were analyzed using logistic regression. The median value of SNHG15 expression was selected as the cut-off value. P<0.05 was considered statistically significant.

RESULTS

Patient Characteristics

The records of 80 primary UM with both RNA-Seq gene expression level and clinicopathological characteristics were obtained from TCGA database. The mean age of 80 UM patients was 61.65 years old, including 45 males and 35 females. The mean value of tumor diameter and thickness were 16.93 and 10.42 mm respectively. In our study cohort, the pathological type of UM included epithelioid cell dominant type and spindle cell dominant type: 42.5% of tumors were epithelioid cell dominant, and 57.5% were spindle cell dominant. There were 68 (85%) cases without extrascleral extension and 7 (8.75%) cases with extrascleral extension. The cancer status included 61 (76.25%) tumor-free cases and 18 (2.25%) cases with tumor. Pathologic stage II was found in 39 (48.75%) cases, and stage III&IV in 40 (50%) cases. And 27 of 80 (33.75%) cases had metastases, 53 of 80 (66.25%) cases had no metastases. Of 80 cases, 23 (28.75%) cases died of all causes.

Survival Outcomes and Multivariate Analysis

Prognostic factors of UM were analyzed using univariate and multivariate Cox regression. The univariate analysis suggested that high SNHG15 expression was a risk factor for death from all causes. Other clinicopathologic variables related to poor prognosis included age, tumor diameter, pathological type, extrascleral extension, cancer status (Table 1). In a multivariate analysis, SNHG15 was an independent risk factor for death from all causes, as was age and pathological type.

Table 1. Prognostic parameters in UM were analyzed using univariate and multivariate Cox regression.

Parameters n/mean Death from all causes
Univariate analysis
Multivariate analysis
P HR 95%CI P HR 95%CI
Age, y 61.65 0.019 1.046 1.008-1.085 0.005 1.064 1.018-1.111
Gender 0.325 0.649 0.274-1.536
 Female 35
 Male 45
Tumor diameter (mm) 16.93 0.034 1.167 1.011-1.347
Thickness (mm) 10.42 0.183 1.118 0.949-1.317
Pathological type 0.001 2.133 1.347-3.379 0.006 1.929 1.212-3.068
 Epithelioid cell dominant 34
 Spindle cell dominant 46
Extrascleral extension 0.008 0.215 0.070-0.667
 No 68
 Yes 7
Cancer status 0.000 0.118 0.050-0.281
 Tumor free 61
 With tumor 18
Pathological stage 0.360 0.666 0.279-1.589
 II 39
 III&IV 40
SNHG15 0.029 0.594 0.373-0.947 0.011 0.525 0.319-0.864
 High 40
 Low 40

UM: Uveal melanoma.

SNHG15 Expression Associated with Clinical Pathological Characteristics

A total of 80 UM cases with SNHG15 expression data and clinicopathologic characteristics were analyzed from TCGA. Kaplan-Meier survival analysis demonstrated that high SNHG15 expression group had a worse prognosis when compared to low SNHG15 expression group (Figure 1A, P<0.05). As shown in Figure 1B-1D, SNHG15 was statistically differentially expressed in diverse groups of the tumor pathologic stage (stage II vs III&IV, P=0.0257), metastasis (P=0.0071), living status (P=0.0017). To clarify the clinicopathologic impact of SNHG15, we also used logistic regression and concluded that the SNHG15 expression (based on median value of 7.94 FPKM) as a categorical variable was statistically related to clinicopathologic features (Table 2). High SNHG15 expression was significantly related to cancer status, pathologic stage, metastasis, living status in UM (all P<0.05; Table 2). These results demonstrated that UM with high SNHG15 expression were prone to progress to cancer status of survival with tumor, a more advanced stage, metastasis and poor living status when compared to the low SNHG15 expression group. However, there was no statistically significant difference in age, gender, tumor diameter, thickness, pathological type, extrascleral invasion.

Figure 1. The SNHG15 expression was associated with clinical pathological characteristics.

Figure 1

A: Patients with high SNHG15 expression had a shorter OS when compared with the low SNHG15 expression group (P=0.02); B-D: The expression of SNHG15 was statistically different in diverse groups of the tumor pathologic stage (P=0.0257), metastasis (P=0.0071), living status (P=0.0017). aP<0.05, bP<0.01.

Table 2. Association between SNHG15 expression and clinicopathologic variables using logistic regression.

Parameters n/mean SNHG15 expression
P OR 95%CI
High Low
Age, y 61.65 62.68 60.63 0.509 1.011 0.979-1.043
Gender 0.499 1.357 0.559-3.292
 Female 35 19 16
 Male 45 21 24
Tumor diameter (mm) 16.93 17.56 16.31 0.113 1.115 0.975-1.275
Thickness (mm) 10.42 10.89 9.95 0.135 1.132 0.962-1.332
Pathological type 0.651 1.227 0.505-2.982
 Epithelial 34 18 16
 Non-epithelial 46 22 24
Extrascleral invasion 0.720 0.750 0.156-3.607
 No 68 34 34
 Yes 7 4 3
Cancer status 0.013 0.212 0.063-0.720
 Tumor free 61 26 35
 With tumor 18 14 4
Pathological stage 0.020 0.336 0.135-0.839
 II 39 14 25
 III&IV 40 25 15
Metastases 0.036 0.355 0.135-0.935
 No 53 22 31
 Yes 27 18 9
Living status 0.009 0.239 0.082-0.696
 Alive 57 23 34
 Dead 23 17 6

Main Enriched Pathways in UM Tissues with High SNHG15 Expression

To explore the SNHG15-related potential signaling pathways activated in UM, GSEA was performed. In the current study, based on the association with SNHG15 expression, the gene list was generated firstly by GSEA. To clarify the statistically significant differences between high and low SNHG15 expression groups, GSEA was conducted subsequently. The results indicated that there were significant differences in spliceosome, cell cycle, pyrimidine metabolism, DNA replication, nucleotide excision repair, RNA degradation, homologous recombination and mismatch repair among patients with high SNHG15 expression phenotype (Figure 2, Table 3).

Figure 2. Enrichment plots from GSEA.

Figure 2

Spliceosome, cell cycle, pyrimidine metabolism, DNA replication, nucleotide excision repair, RNA degradation, homologous recombination and mismatch repair are enriched significantly in SNHG15 high expression phenotype.

Table 3. Enriched pathways for differential SNHG15 expression in UM.

Name ES NES Nominal P-val FDR Q-val
Spliceosome 0.606114 1.607263 0.018256 0.151837
Cell cycle 0.651762 1.670409 0.002028 0.148961
Pyrimidine metabolism 0.581931 1.669619 0.014113 0.113017
DNA replication 0.689555 1.597633 0.030928 0.151044
Nucleotide excision repair 0.675713 1.704181 0 0.129225
RNA degradation 0.663434 1.628078 0.003992 0.179965
Homologous recombination 0.759477 1.724602 0 0.182268
Mismatch repair 0.7302 1.617004 0.01222 0.154619

UM: Uveal melanoma; ES: Enrichment score; NES: Normal enrichment score; FDR: False discovery rate.

DISCUSSION

Accumulating evidences indicate that SNHG15 plays a dual role in the tumorigenesis and development of different tumors[26]. Previously, SNHG15 has been demonstrated as a carcinogenic lncRNA, which is usually upregulated in tumor tissues compared with normal tissues[19],[27]. It exerts an oncogenic effect via various epigenetic mechanisms[17],[28]. For example, it can suppress the expression of miR-338-3p and facilitate the proliferation of colorectal cancer cells[29]. It plays a carcinogenic role by affecting miR-338-3p/FKBP1A axis in prostate cancer[23]. It can also enhance hepatocellular carcinoma progression by negative regulation of miR-141-3p[21]. However, there are reports that SNHG15 has a tumor suppressive effect, suggesting that low SNHG15 expression is related to poor prognosis in thyroid cancer and upregulating expression of SNHG15 can significantly suppress cell proliferation[30][31]. At present, the impact of SNHG15 on UM is still unclear. Therefore, vital roles and potential biological mechanism of SNHG15 in UM needs to be elucidated.

In this study, we revealed that high SNHG15 expression was related to clinicopathologic features in UM. Through RNA-Seq gene expression level and clinicopathological characteristics obtained from the TCGA UM project, we analyzed the relationship among SNHG15 expression, clinicopathological features and prognosis of UM. The univariate analysis demonstrated that SNHG15 expression level, age, tumor diameter, pathological type, extrascleral extension, and cancer status were risk factors for death from all causes. The multivariate analysis suggested that high SNHG15 expression along with age and pathological type was an independent risk factor for death from all causes. Therefore, the results demonstrated that high SNHG15 expression was an independent predictor of poor prognosis in UM through univariate and multivariate analysis.

Kaplan-Meier survival analysis also indicated that high SNHG15 expression group had a worse prognosis when compared to low SNHG15 expression group in UM. In addition, an analysis was conducted to further explore the relationship between SNHG15 and clinicopathological features. The SNHG15 expression was statistically different in diverse groups of the tumor pathologic stage, metastasis and living status. Besides, high SNHG15 expression (based on median expression value of 7.94 FPKM) in UM was associated with cancer status of survival with tumor, advanced pathologic stage, metastasis and living status. It demonstrated that high SNHG15 expression in UM was strongly related to poor prognosis.

The mechanisms of SNHG15 dysregulation in malignant tumors are quite complex and are far from being completely understood. Previous studies have suggested that SNHG15 is involved in diverse pathological and physiological processes of many tumors through their abnormal expressions, including cell proliferation, invasion, migration and autophagy[17],[29]. To explore the biological mechanism of SNHG15 in UM, GSEA was conducted. It indicated that spliceosome, cell cycle, pyrimidine metabolism, DNA replication, nucleotide excision repair, RNA degradation, homologous recombination and mismatch repair were all enriched differentially in SNHG15 high expression phenotype. Alternative splicing is essential for gene regulation, and abnormal splicing plays a vital role in inactivating tumor suppressor genes or activating oncogenes[32]. SNHG15 may have an impact on the invasion and migration of UM cells by affecting spliceosomal related factors. The abnormal cell proliferation of tumor is related to the lack of checkpoint control over the cell cycle, which is the basis of genetic instability[33]. Evidence shows that the lack of homologous recombination may facilitate the disturbance of cell cycle, the instability and accumulated mutations of genome during the progression and development[34]. Mismatch repair proteins have an significant role in DNA hypermethylation alteration and tumorigenesis[35]. SNHG15 is closely related to DNA replication and mismatch repair, demonstrating that SNHG15 may promote the occurrence of UM by affecting DNA replication and DNA mismatch repair. It indicated that SNHG15 may be identified as a novel marker of diagnosis, therapeutic and prognosis prediction in UM. However, the related mechanism needs to be further elucidated.

This research also has some limitations. The most important one is the limited number of patients and time of follow-up. In addition, some patient characteristics (such as ciliary body involvement) were not completely recorded in the database. In fact, ciliary body involvement plays a critical role in UM[36][37].

In conclusion, this study aims to demonstrate the vital role of SNHG15 in UM and the potential relationship between SNHG15 expression and clinical parameters. SNHG15 expression may be a valuable biomarker for poor survival in UM. Moreover, we have preliminarily explored the crucial pathway associated with SNHG15 in UM. However, further experimental validation is needed to be performed for clarifying the significant impact of SNHG15. And it is of great significance to further identify its independent prognostic value in a large-scale, standardized researches on UM.

Acknowledgments

Foundations: Supported by the National Natural Science Foundation of China (No.81970835; No.81800867).

Conflicts of Interest: Wu X, None; Li XF, None; Wu Q, None; Ma RQ, None; Qian J, None; Zhang R, None.

REFERENCES

  • 1.van Raamsdonk CD, Griewank KG, Crosby MB, Garrido MC, Vemula S, Wiesner T, Obenauf AC, Wackernagel W, Green G, Bouvier N, Sozen MM, Baimukanova G, Roy R, Heguy A, Dolgalev I, Khanin R, Busam K, Speicher MR, O'Brien J, Bastian BC. Mutations in GNA11 in uveal melanoma. N Engl J Med. 2010;363(23):2191–2199. doi: 10.1056/NEJMoa1000584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Carvajal RD, Sosman JA, Quevedo JF, Milhem MM, Joshua AM, Kudchadkar RR, Linette GP, Gajewski TF, Lutzky J, Lawson DH, Lao CD, Flynn PJ, Albertini MR, Sato T, Lewis K, Doyle A, Ancell K, Panageas KS, Bluth M, Hedvat C, Erinjeri J, Ambrosini G, Marr B, Abramson DH, Dickson MA, Wolchok JD, Chapman PB, Schwartz GK. Effect of selumetinib vs chemotherapy on progression-free survival in uveal melanoma: a randomized clinical trial. JAMA. 2014;311(23):2397–2405. doi: 10.1001/jama.2014.6096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Shain AH, Bagger MM, Yu R, Chang D, Liu SS, Vemula S, Weier JF, Wadt K, Heegaard S, Bastian BC, Kiilgaard JF. The genetic evolution of metastatic uveal melanoma. Nat Genet. 2019;51(7):1123–1130. doi: 10.1038/s41588-019-0440-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bagger M, Smidt-Nielsen I, Andersen MK, Jensen PK, Heegaard S, Andersen KK, Friis S, Kiilgaard JF. Long-term metastatic risk after biopsy of posterior uveal melanoma. Ophthalmology. 2018;125(12):1969–1976. doi: 10.1016/j.ophtha.2018.03.047. [DOI] [PubMed] [Google Scholar]
  • 5.Kujala E, Mäkitie T, Kivelä T. Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci. 2003;44(11):4651–4659. doi: 10.1167/iovs.03-0538. [DOI] [PubMed] [Google Scholar]
  • 6.Chandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, Wunderlich JR, Danforth DN, Zlott D, Paria BC, Sabesan AC, Srivastava AK, Xi LQ, Pham TH, Raffeld M, White DE, Toomey MA, Rosenberg SA, Kammula US. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017;18(6):792–802. doi: 10.1016/S1470-2045(17)30251-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mendell JT. Targeting a long noncoding RNA in breast cancer. N Engl J Med. 2016;374(23):2287–2289. doi: 10.1056/NEJMcibr1603785. [DOI] [PubMed] [Google Scholar]
  • 8.Lan Y, Xiao XW, He ZC, Luo Y, Wu CF, Li L, Song X. Long noncoding RNA OCC-1 suppresses cell growth through destabilizing HuR protein in colorectal cancer. Nucleic Acids Res. 2018;46(11):5809–5821. doi: 10.1093/nar/gky214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cao CH, Sun JY, Zhang DY, Guo XJ, Xie LW, Li X, Wu DH, Liu L. The long intergenic noncoding RNA UFC1, a target of microRNA 34a, interacts with the mRNA stabilizing protein HuR to increase levels of β-catenin in HCC cells. Gastroenterology. 2015;148(2):415–426.e18. doi: 10.1053/j.gastro.2014.10.012. [DOI] [PubMed] [Google Scholar]
  • 10.Wang P, Xue YQ, Han YM, Lin L, Wu C, Xu S, Jiang ZP, Xu JF, Liu QY, Cao XT. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science. 2014;344(6181):310–313. doi: 10.1126/science.1251456. [DOI] [PubMed] [Google Scholar]
  • 11.Zheng XL, Tang HW, Zhao XF, Sun YM, Jiang YF, Liu YH. Long non-coding RNA FTH1P3 facilitates uveal melanoma cell growth and invasion through miR-224-5p. PLoS One. 2017;12(11):e0184746. doi: 10.1371/journal.pone.0184746. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 12.Lu QK, Zhao N, Zha GP, Wang HY, Tong QH, Xin SH. LncRNA HOXA11-AS exerts oncogenic functions by repressing p21 and miR-124 in uveal melanoma. DNA Cell Biol. 2017;36(10):837–844. doi: 10.1089/dna.2017.3808. [DOI] [PubMed] [Google Scholar]
  • 13.Lu LN, Yu XY, Zhang LL, Ding X, Pan H, Wen XY, Xu SQ, Xing Y, Fan JY, Ge SF, Zhang H, Jia RB, Fan XQ. The long non-coding RNA RHPN1-AS1 promotes uveal melanoma progression. Int J Mol Sci. 2017;18(1):226. doi: 10.3390/ijms18010226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wu S, Chen H, Han N, Zhang CX, Yan HT. Long noncoding RNA PVT1 silencing prevents the development of uveal melanoma by impairing MicroRNA-17-3p-dependent MDM2 upregulation. Invest Ophthalmol Vis Sci. 2019;60(14):4904–4914. doi: 10.1167/iovs.19-27704. [DOI] [PubMed] [Google Scholar]
  • 15.Li P, He J, Yang Z, Ge SF, Zhang H, Zhong Q, Fan XQ. ZNNT1 long noncoding RNA induces autophagy to inhibit tumorigenesis of uveal melanoma by regulating key autophagy gene expression. Autophagy. 2019:1–14. doi: 10.1080/15548627.2019.1659614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Dong YZ, Meng XM, Li GS. Long non-coding RNA SNHG15 indicates poor prognosis of non-small cell lung cancer and promotes cell proliferation and invasion. Eur Rev Med Pharmacol Sci. 2018;22(9):2671–2679. doi: 10.26355/eurrev_201805_14963. [DOI] [PubMed] [Google Scholar]
  • 17.Liu K, Hou Y, Liu YK, Zheng J. LncRNA SNHG15 contributes to proliferation, invasion and autophagy in osteosarcoma cells by sponging miR-141. J Biomed Sci. 2017;24(1):46. doi: 10.1186/s12929-017-0353-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wu DM, Wang S, Wen X, Han XR, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ, Hu B, Lu J, Chen GQ, Zheng YL. LncRNA SNHG15 acts as a ceRNA to regulate YAP1-Hippo signaling pathway by sponging miR-200a-3p in papillary thyroid carcinoma. Cell Death Dis. 2018;9(10):947. doi: 10.1038/s41419-018-0975-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Guo XB, Yin HS, Wang JY. Evaluating the diagnostic and prognostic value of long non-coding RNA SNHG15 in pancreatic ductal adenocarcinoma. Eur Rev Med Pharmacol Sci. 2018;22(18):5892–5898. doi: 10.26355/eurrev_201809_15917. [DOI] [PubMed] [Google Scholar]
  • 20.Sun XT, Bai Y, Yang C, Hu SY, Hou ZL, Wang GX. Long noncoding RNA SNHG15 enhances the development of colorectal carcinoma via functioning as a ceRNA through miR-141/SIRT1/Wnt/β-catenin axis. Artif Cells Nanomed Biotechnol. 2019;47(1):2536–2544. doi: 10.1080/21691401.2019.1621328. [DOI] [PubMed] [Google Scholar]
  • 21.Ye JF, Tan LD, Fu Y, Xu HJ, Wen LJ, Deng Y, Liu K. LncRNA SNHG15 promotes hepatocellular carcinoma progression by sponging miR-141-3p. J Cell Biochem. 2019;120(12):19775–19783. doi: 10.1002/jcb.29283. [DOI] [PubMed] [Google Scholar]
  • 22.Zhang JH, Wei HW, Yang HG. Long noncoding RNA SNHG15, a potential prognostic biomarker for hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 2016;20(9):1720–1724. [PubMed] [Google Scholar]
  • 23.Zhang YL, Zhang DH, Lv J, Wang S, Zhang Q. LncRNA SNHG15 Acts as an oncogene in prostate cancer by regulating miR-338-3p/FKBP1A axis. Gene. 2019;705:44–50. doi: 10.1016/j.gene.2019.04.033. [DOI] [PubMed] [Google Scholar]
  • 24.Kong QL, Qiu M. Long noncoding RNA SNHG15 promotes human breast cancer proliferation, migration and invasion by sponging miR-211-3p. Biochem Biophys Res Commun. 2018;495(2):1594–1600. doi: 10.1016/j.bbrc.2017.12.013. [DOI] [PubMed] [Google Scholar]
  • 25.Wang TQ, Sun HB, Bao Y, En R, Tian YJ, Zhao W, Jia LZ. POM121 overexpression is related to a poor prognosis in colorectal cancer. Expert Rev Mol Diagn. 2020;20(3):345–353. doi: 10.1080/14737159.2020.1707670. [DOI] [PubMed] [Google Scholar]
  • 26.Shuai Y, Ma ZH, Lu JW, Feng JF. LncRNA SNHG15: a new budding star in human cancers. Cell Prolif. 2020;53(1):e12716. doi: 10.1111/cpr.12716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Qu C, Dai CM, Guo YH, Qin R, Liu JB. Long noncoding RNA SNHG15 serves as an oncogene and predicts poor prognosis in epithelial ovarian cancer. Onco Targets Ther. 2019;12:101–111. doi: 10.2147/OTT.S182657. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 28.Ma YW, Xue YX, Liu XB, Qu CB, Cai H, Wang P, Li ZQ, Li Z, Liu YH. SNHG15 affects the growth of glioma microvascular endothelial cells by negatively regulating miR-153. Oncol Rep. 2017;38(5):3265–3277. doi: 10.3892/or.2017.5985. [DOI] [PubMed] [Google Scholar]
  • 29.Li M, Bian ZH, Jin GY, Zhang J, Yao SR, Feng YY, Wang X, Yin Y, Fei BJ, You QJ, Huang ZH. LncRNA-SNHG15 enhances cell proliferation in colorectal cancer by inhibiting miR-338-3p. Cancer Med. 2019;8(5):2404–2413. doi: 10.1002/cam4.2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Liu YC, Li JL, Li F, Li M, Shao Y, Wu LP. SNHG15 functions as a tumor suppressor in thyroid cancer. J Cell Biochem. 2019;120(4):6120–6126. doi: 10.1002/jcb.27899. [DOI] [PubMed] [Google Scholar]
  • 31.Liu YC, Li JL, Li M, Li F, Shao Y, Wu LP. microRNA-510-5p promotes thyroid cancer cell proliferation, migration, and invasion through suppressing SNHG15. J Cell Biochem. 2019 doi: 10.1002/jcb.28454. [DOI] [PubMed] [Google Scholar]
  • 32.Li YW, Guo HY, Jin CJ, Qiu CP, Gao M, Zhang L, Liu ZJ, Kong BH. Spliceosome-associated factor CTNNBL1 promotes proliferation and invasion in ovarian cancer. Exp Cell Res. 2017;357(1):124–134. doi: 10.1016/j.yexcr.2017.05.008. [DOI] [PubMed] [Google Scholar]
  • 33.Williams GH, Stoeber K. The cell cycle and cancer. J Pathol. 2012;226(2):352–364. doi: 10.1002/path.3022. [DOI] [PubMed] [Google Scholar]
  • 34.Yu B, Ding YM, Liao XF, Wang CH, Wang B, Chen XY. Overexpression of PARPBP correlates with tumor progression and poor prognosis in hepatocellular carcinoma. Dig Dis Sci. 2019;64(10):2878–2892. doi: 10.1007/s10620-019-05608-4. [DOI] [PubMed] [Google Scholar]
  • 35.Maiuri AR, Peng M, Podicheti R, Sriramkumar S, Kamplain CM, Rusch DB, DeStefano Shields CE, Sears CL, O'Hagan HM. Mismatch repair proteins initiate epigenetic alterations during inflammation-driven tumorigenesis. Cancer Res. 2017;77(13):3467–3478. doi: 10.1158/0008-5472.CAN-17-0056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Berry D, Seider M, Stinnett S, Mruthyunjaya P, Schefler AC, Ocular Oncology Study Consortium Relationship of clinical features and baseline tumor size with gene expression profile status in uveal melanoma: a Multi-institutional study. Retina. 2019;39(6):1154–1164. doi: 10.1097/IAE.0000000000002113. [DOI] [PubMed] [Google Scholar]
  • 37.Jiang ZM, Yu FH, Li M. Upregulation of BCL2 19 kD protein-interacting protein 3 (BNIP3) is predictive of unfavorable prognosis in uveal melanoma. Med Sci Monit. 2018;24:4711–4717. doi: 10.12659/MSM.907679. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Ophthalmology are provided here courtesy of Press of International Journal of Ophthalmology

RESOURCES