A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma

  • Authors:
    • Qiong Yi
    • Wen‑Jin Zou
  • View Affiliations

  • Published online on: December 14, 2018     https://doi.org/10.3892/mmr.2018.9766
  • Pages: 1294-1301
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Uveal melanoma (UM), the predominant histological subtype of intraocular malignant tumors in adults, often results in high rates of mortality; effective prognostic signatures used to predict the survival of patients with UM are limited. Small nucleolar RNAs (snoRNAs) are emerging as important regulators in the processes of carcinogenesis and tumor progression, but knowledge of their application as prognostic markers in UM is limited. In the present study, the expression profiles of snoRNAs in UM were determined; a total of 60 snoRNAs were notably associated with the overall survival of patients with UM via univariate Cox survival analysis. Subsequently, a prognostic signature based on four snoRNAs was proposed, which retained their prognostic significance determined by a multivariate Cox survival analysis. The formula is as follows: ACA17 * (‑1.602) + ACA45 * 0.803 + HBII‑276 * 0.603 + SNORD12 * 1.348. Furthermore, the results of in silico analysis indicated that perturbation of the phototransduction, GABAergic synapse and amphetamine addiction pathways may be the potential molecular mechanisms underlying the poor prognosis of patients with UM. Collectively, the present study proposed a potential prognostic signature for patients with UM and the prospective mechanisms at the genome‑wide level were determined.

Introduction

Uveal melanoma (UM), a rare subset of melanoma accounting for only 5% of all melanomas, makes up 85–95% of all ocular melanomas; however, UM ranks as the predominant intraocular malignancy in adult patients (14). Tumors located on the choroid are the most common, affecting ≤90% of patients (57). In addition, ~50% of patients will develop metastasis, >90% of which disseminates to the liver (8,9). Once the tumor has metastasized, the median overall survival (OS) is only 6–12 months (1012). The average age at diagnosis is 60-years-old, and UM affects both sexes almost equally, with a slightly increased frequency in males (13,14). Recent advances in the detection of molecular pathology and a various immune-based therapies have exhibited efficacy in treating UM. However, the molecular biomarkers that may be effective in predicting the development and prognosis of UM in patients are limited, which has led to the unsatisfactory clinical management and targeted therapy of this disease (1517); thus, UM is associated with high mortality. Therefore, there is an urgent need to identify ideal prognostic biomarkers and treatment options for patients with UM.

Recently, increasing evidence has indicated that non-coding RNAs serve a vital regulatory role in tumorigenesis and tumor progression, including small nucleolar RNAs (snoRNAs) (18,19). snoRNAs, encoded by introns, exist stably in the cellular environment and can regulate the expression of specific genes (2023). Numerous studies have proposed that snoRNA expression profiles are dysregulated in various types of tumors (24,25). Furthermore, numerous dysregulated snoRNAs have also been identified to be associated with the development and prognosis of cancer, including melanoma, which suggests the potential of snoRNAs as prognostic predictors (2628). Unfortunately, the association between snoRNAs and UM has rarely been reported.

In the present study, the expression landscape of snoRNAs was integrated using the snoRNA in cancers (SNORic) database and the clinical information of patients with UM in The Cancer Genome Atlas (TCGA) database. snoRNas associated with survival were systematically selected and a specific prognosis index (PI) was proposed, which could be an ideal prognostic signature to predict the prognosis of patients with UM. Further bioinformatics analysis revealed survival-associated pathways in UM. These findings may aid the identification of the novel biological functions of snoRNAs in UM, and improve the clinical management of this disease.

Materials and methods

General characteristics

The data of 80 patients with UM were submitted for survival analysis. Among the 80 patients with UM, the histological types included epithelioid cell (n=34) and spindle cell (n=46). A total of 35 patients were female and 45 patients were male. The average age was 61.65 years (ranging from 22–86 years).

Data acquisition

SnoRNA expression profiles were downloaded from the SNORic (http://bioinfo.life.hust.edu.cn/SNORic) database, which provides the quantified expression levels of snoRNAs in the form of reads per kilobase per million (RPKM). Then, the original values were calculated via log2 conversion. The corresponding clinical information of UM patients was also downloaded from the TCGA data portal (http://cancergenome.nih.gov/). To obtain more accurate results, low-abundance snoRNAs with average log (RPKM+1) expression levels in all samples were omitted.

Survival analysis

A univariate Cox regression analysis was performed to select snoRNAs that were correlated with the overall survival (OS) of patients with UM. Subsequently, multivariate Cox proportional hazards regression was implemented to further identify whether the prognostic snoRNAs were independent biomarkers. Finally, the snoRNA-based prognostic signature was proposed according to the linear multiplication of the expression profiles of each prognostic snoRNA, weighted by their estimated regression coefficients in the multivariate Cox analysis. All survival analyses were conducted using the Survival package version 2.43-1 in R software version 3.4.4 (https://CRAN.R-project.org/package=survival). Then, patients were separated into high- and low-risk groups according to median risk score values. Kaplan-Meier (K-M) survival analysis followed by a log-rank test was used to assess the value of the snoRNAs in predicting prognosis. The number of patients in the low- and high-risk groups was recorded every 500 days.

Bioinformatics procedure

To further investigate the potential pathways affected in the high- and low-risk groups, the Limma package version 3.38.2 in R software was used to identify the differentially expressed genes between high- and low-risk groups (29). The mRNA expression profiles were downloaded from TCGA database; the raw count data of mRNAs were submitted for analysis. The thresholds for definite differentially expressed genes were set to |log fold change|>2 and false discovery rate <0.01. Subsequently, differentially expressed genes were submitted to the DAVID online database for gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/) analysis. GO analysis includes three categories: ‘Biological process’ (BP), ‘cellular component’ (CC) and ‘molecular function’ (MF). Furthermore, a protein-protein interaction (PPI) network was also generated to demonstrate the association between these genes. PPI analysis was performed using STRING version 10.5 (https://string-db.org/). The results were visually presented using the ggplot2 package version 3.1.0 in R software (30). Providing that BRCA1-associated protein-1 (BAP1) mutation is a critical indicator for monitoring the poor progression of UM, the prognostic value of BAP1 mutation was determined in the present study. Data for UM patients, including BAP1 mutation status, were downloaded from the cBioportal database (http://www.cbioportal.org/).

Results

Survival-associated snoRNAs

After removing the low-abundance snoRNAs, 380 snoRNAs were submitted for survival analysis. Among these, 64 snoRNAs were significantly associated with the OS of patients with UM (P<0.05). The top 20 most significant survival-associated snoRNAs were presented in Fig. 1. Then, multivariate Cox analysis was performed and four snoRNAs were selected to construct a prognostic signature, including ACA45 (chr15: 83424696_83424823), ACA17 (chr9: 139621198_139621331), HBII-276 (chr8: 67834708_67834784) and SNORD12 (chr20: 47897219_47897309). A survival-risk formula was constructed based on the four snoRNAs as follows: ACA17 * (−1.602) + ACA45 * 0.803 + HBII-276 * 0.603 + SNORD12 * 1.348. Then, patients were divided into high- and low-risk groups according to median value of prognostic signature (Fig. 2). K-M survival analysis revealed that patients in the high-risk group exhibited shorter survival than those patients in the low-risk group (Fig. 3A). The number of patients in the high- and low-risk groups, and the number of censored patients at different time points are shown in Fig. 3B and C. ACA17 was significantly upregulated in the low-risk group, while ACA45, HBII-276 and SNORD12 were significantly upregulated in the high-risk group (Fig. 4). K-M survival plots were also used to present the prognostic value of each snoRNA (Fig. 5). ACA17 upregulation indicated better clinical outcome, while ACA45, HBII-276 and SNORD12 upregulation was associated with the poor survival of patients with UM.

Functional enrichment analysis

A total of 281 differentially expressed mRNAs between the high- and low-risk groups were determined (Fig. 6). PPI network analysis suggested that these genes notably interacted with each other (Fig. 7). Gene functional enrichment analysis revealed that these genes were significantly enriched in several biological processes and pathways. For BP, the most notably enriched functional terms were ‘visual perception’, ‘regulation of rhodopsin-mediated signaling pathway’ and ‘rhodopsin-mediated signaling pathway’ (Fig. 8A). Regarding CC, genes were markedly enriched in ‘photoreceptor inner segment’, ‘photoreceptor disc membrane’ and ‘photoreceptor outer segment’ (Fig. 8B). In regards to MF, genes were notably enriched in ‘structural constituent of eye lens’, ‘structural molecule activity’ and ‘hormone activity’ (Fig. 8C). Importantly, KEGG enrichment analysis suggested that these genes were significantly associated with ‘phototransduction’, ‘γ-aminobutyric acid (GABA) ergic synapse’, ‘amphetamine (APA) addiction’, ‘nicotine addiction’ and ‘neuroactive ligand-receptor interaction’ (Fig. 9). Furthermore, the mutation status of BAP1 may be useful in predicting the clinical outcome of patients with UM; mutations in BAP1 demonstrated a significant association with poorer overall survival compared with UM patients without BAP1 mutations (Fig. 10A). Of note, the prognostic signature based on snoRNAs was markedly associated high-risk patients with BAP1 mutations than those without in the low-risk group (Fig. 10B).

Discussion

In the present study, survival-associated snoRNAs in patients with UM were selected. Then, the snoRNAs that exhibited a significant prognostic value were determined and identified by multivariate analysis. Most importantly, a prognostic signature comprising four novel snoRNAs was proposed: ACA17, ACA45, HBII276 and SNORD12. Notably, the prognostic signature of the present study, based on these four novel snoRNAs, may be an ideal risk model for patients with UM. To the best of our knowledge, the present study is the first to propose a prognostic signature based on snoRNAs. Furthermore, gene functional enrichment analysis revealed that the optic nerve-associated pathways are dysregulated most significantly between high-risk and low-risk groups. These findings may provide novel insight into the clinical management and molecular mechanisms underlying UM.

Previously, few studies have investigated the PI of UM from TCGA database. Xu et al (31) reported that high plasmacytoma variant translocation 1 (PVT1) expression was an independent predictor in patients with UM; it was inferred that the expression of PVT1 may act as a moderate and specific prognostic factor in terms of OS in UM. Wan et al (32) identified 21 co-expression modules that were analyzed based on 10,975 genes from 80 UM samples, by using weighted correlation network analysis. It was revealed that the hub genes solute carrier family 17 member 7, neurotrophic receptor tyrosine kinase 2, ankyrin repeat and BTB domain containing 1 and ADP-ribosylhydrolase like 1 may also serve a role as potential diagnostic and prognostic biomarkers of UM. Robertson et al (33) also analyzed the genomic data of 80 patients with UM; the patients were divided into several subgroups with different genomic aberrations, transcriptional features and clinical outcomes. Field et al (34) not only identified preferentially expressed antigen in melanoma (PRAME) expression as a biomarker for increased metastatic risk in class 1 UM; however, PRAME expression was associated with poor prognosis among class 2 UM cases (35). These studies provided novel and constructive methods of selecting prognostic biomarkers for UM; however, the role of snoRNAs in the prognosis of UM remains unclear. In the present study, a novel prognostic signature based on snoRNAs was developed. More importantly, the proposed risk score may attain satisfactory prognostic values for UM. With the advantage of high-throughput RNA-sequencing, numerous snoRNAs were identified; however, investigations into the clinical significance of snoRNAs in UM are limited. The present study may provide novel insight into the clinical management of UM.

Gene functional enrichment analysis revealed that several biological processes and pathways were associated with differentially expressed mRNAs between the high- and low-risk groups; however, the biological mechanisms underlying this association are unclear. The genes detected from KEGG enrichment analysis were demonstrated to be associated with ‘phototransduction’, ‘GABAergic synapse’ and ‘APA addiction’ in particular. Brown et al (36) revealed that brain-modulated choroidal thickness has an unusual and well-established light dependence. In addition, Wicks et al (37) reported that ultraviolet phototransduction can evoke retina-dependent calcium flux in human epidermal melanocytes, and increased cellular melanin content. Furthermore, ocular melanocytosis is an important predisposing factor for UM (38). Therefore, it was speculated that phototransduction may be involved in the formation of UM in the present study. GABAergic synapses are important inhibitory neurotransmitters in the mammalian central nervous system (CNS), as they serve to hyperpolarize the postsynaptic neuron (39). Interestingly, ‘APA addiction’, ‘nicotine addiction’ and ‘dopaminergic (DA) synapses’ were also associated with UM; however, DA is an excitatory neurotransmitter in the CNS (40). APA promotes DA release in the CNS, whilst also inducing other biogenic amine-releasing and inhibitory neurons, and vesicle single-amine transporters (41). Monoamine oxidase can excite the CNS (42). Nicotine initially inhibits DA release, which is then enhanced by regulating GABAergic neurons (43). This mechanism of action is similar to that of APAs in the CNS. Thus, it may be inferred that CNS activity-associated pathways may be notably dysregulated between high-risk and low-risk groups.

However, several limitations should be considered. An additional independent cohort was not used to validate the performance of prognostic signature. Furthermore, the in silico analysis for the molecular mechanisms require further investigation.

Collectively, the results of the present study demonstrated that snoRNAs may be notable prognostic markers for the survival of patients with UM. To the best of our knowledge, the present study is the first to demonstrate the prognostic value of snoRNAs, and provides novel insight into the complex biological functions underlying UM.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81160119), a funding of Natural Science Foundation of Guangxi Zhuang Autonomous Region (grant nos. 2017GXNSFAA198347 and 2016GXNsFAA380280).

Availability of data and materials

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

Authors' contributions

W-JZ made substantial contributions to the design of the present study. QY and W-JZ conducted statistical analysis. QY wrote the manuscript which was revised by W-JZ. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Álvarez-Rodríguez B, Latorre A, Posch C and Somoza Á: Recent advances in uveal melanoma treatment. Med Res Rev. 37:1350–1372. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Komatsubara KM, Manson DK and Carvajal RD: Selumetinib for the treatment of metastatic uveal melanoma: Past and future perspectives. Future Oncol. 12:1331–1344. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Reichstein D: New concepts in the molecular understanding of uveal melanoma. Curr Opin Ophthalmol. 28:219–227. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Vasalaki M, Fabian ID, Reddy MA, Cohen VM and Sagoo MS: Ocular oncology: Advances in retinoblastoma, uveal melanoma and conjunctival melanoma. Br Med Bull. 121:107–119. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Kaliki S and Shields CL: Uveal melanoma: Relatively rare but deadly cancer. Eye (Lond). 31:241–257. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Shields CL, Kels JG and Shields JA: Melanoma of the eye: Revealing hidden secrets, one at a time. Clin Dermatol. 33:183–196. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Helgadottir H and Höiom V: The genetics of uveal melanoma: Current insights. Appl Clin Genet. 9:147–155. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Heppt MV, Steeb T, Schlager JG, Rosumeck S, Dressler C, Ruzicka T, Nast A and Berking C: Immune checkpoint blockade for unresectable or metastatic uveal melanoma: A systematic review. Cancer Treat Rev. 60:44–52. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Blum ES, Yang J, Komatsubara KM and Carvajal RD: Clinical management of uveal and conjunctival melanoma. Oncology (Williston Park). 30:29–32, 34-43, 48. 2016.PubMed/NCBI

10 

Krantz BA, Dave N, Komatsubara KM, Marr BP and Carvajal RD: Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clin Ophthalmol. 11:279–289. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zhou J, Jin B, Jin Y, Liu Y and Pan J: The antihelminthic drug niclosamide effectively inhibits the malignant phenotypes of uveal melanoma in vitro and in vivo. Theranostics. 7:1447–1462. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Cheng H, Chua V, Liao C, Purwin TJ, Terai M, Kageyama K, Davies MA, Sato T and Aplin AE: Co-targeting HGF/cMET signaling with MEK inhibitors in metastatic uveal melanoma. Mol Cancer Ther. 16:516–528. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Mahendraraj K, Lau CS, Lee I and Chamberlain RS: Trends in incidence, survival, and management of uveal melanoma: A population-based study of 7,516 patients from the surveillance, epidemiology, and end results database (1973–2012). Clin Ophthalmol. 10:2113–2119. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Andreoli MT, Mieler WF and Leiderman YI: Epidemiological trends in uveal melanoma. Br J Ophthalmol. 99:1550–1553. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Chandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, Wunderlich JR, Danforth DN, Zlott D, Paria BC, et al: Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: A single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 18:792–802. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Komatsubara KM and Carvajal RD: Immunotherapy for the treatment of uveal melanoma: Current status and emerging therapies. Curr Oncol Rep. 19:452017. View Article : Google Scholar : PubMed/NCBI

17 

Spagnolo F, Picasso V, Spano L, Tanda E, Venzano C and Queirolo P: Update on metastatic uveal melanoma: Progress and challenges. BioDrugs. 30:161–172. 2016. View Article : Google Scholar : PubMed/NCBI

18 

McMahon M, Contreras A and Ruggero D: Small RNAs with big implications: New insights into H/ACA snoRNA function and their role in human disease. Wiley Interdiscip Rev RNA. 6:173–189. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Zhou H, Xu Q, Ni C, Ye S, Xu X, Hu X, Jiang J, Hong Y, Huang D and Yang L: Prospects of noncoding RNAs in hepatocellular carcinoma. Biomed Res Int. 2018:65794362018. View Article : Google Scholar : PubMed/NCBI

20 

Koduru SV, Leberfinger AN and Ravnic DJ: Small non-coding RNA abundance in adrenocortical carcinoma: A footprint of a rare cancer. J Genomics. 5:99–118. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Siprashvili Z, Webster DE, Johnston D, Shenoy RM, Ungewickell AJ, Bhaduri A, Flockhart R, Zarnegar BJ, Che Y, Meschi F, et al: The noncoding RNAs SNORD50A and SNORD50B bind K-Ras and are recurrently deleted in human cancer. Nat Genet. 48:53–58. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Y, Xu C, Gu D, Wu M, Yan B, Xu Z, Wang Y and Liu H: H/ACA box small nucleolar RNA 7A promotes the self-renewal of human umbilical cord mesenchymal stem cells. Stem Cells. 35:222–235. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Huang C, Shi J, Guo Y, Huang W, Huang S, Ming S, Wu X, Zhang R, Ding J, Zhao W, et al: A snoRNA modulates mRNA 3′ end processing and regulates the expression of a subset of mRNAs. Nucleic Acids Res. 45:8647–8660. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Zhou F, Liu Y, Rohde C, Pauli C, Gerloff D, Köhn M, Misiak D, Bäumer N, Cui C, Göllner S, et al: AML1-ETO requires enhanced C/D box snoRNA/RNP formation to induce self-renewal and leukaemia. Nat Cell Biol. 19:844–855. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Xu B, Ye MH, Lv SG, Wang QX, Wu MJ, Xiao B, Kang CS and Zhu XG: SNORD47, a box C/D snoRNA, suppresses tumorigenesis in glioblastoma. Oncotarget. 8:43953–43966. 2017.PubMed/NCBI

26 

Patterson DG, Roberts JT, King VM, Houserova D, Barnhill EC, Crucello A, Polska CJ, Brantley LW, Kaufman GC, Nguyen M, et al: Human snoRNA-93 is processed into a microRNA-like RNA that promotes breast cancer cell invasion. NPJ Breast Cancer. 3:252017. View Article : Google Scholar : PubMed/NCBI

27 

Yoshida K, Toden S, Weng W, Shigeyasu K, Miyoshi J, Turner J, Nagasaka T, Ma Y, Takayama T, Fujiwara T and Goel A: SNORA21-an oncogenic small nucleolar RNA, with a prognostic biomarker potential in human colorectal cancer. EBioMedicine. 22:68–77. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Xu L, Ziegelbauer J, Wang R, Wu WW, Shen RF, Juhl H, Zhang Y and Rosenberg A: Distinct profiles for mitochondrial t-RNAs and small nucleolar RNAs in locally invasive and metastatic colorectal cancer. Clin Cancer Res. 22:773–784. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

30 

Wickham H: ggplot2: Elegant graphics for data analysis. Springer Publishing Company, Incorporated. 2009.

31 

Xu H, Gong J and Liu H: High expression of lncRNA PVT1 independently predicts poor overall survival in patients with primary uveal melanoma. PLoS One. 12:e01896752017. View Article : Google Scholar : PubMed/NCBI

32 

Wan Q, Tang J, Han Y and Wang D: Co-expression modules construction by WGCNA and identify potential prognostic markers of uveal melanoma. Exp Eye Res. 166:13–20. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Robertson AG, Shih J, Yau C, Gibb EA, Oba J, Mungall KL, Hess JM, Uzunangelov V, Walter V, Danilova L, et al: Integrative analysis identifies four molecular and clinical subsets in uveal melanoma. Cancer Cell. 32:1512018. View Article : Google Scholar

34 

Field MG, Decatur CL, Kurtenbach S, Gezgin G, van der Velden PA, Jager MJ, Kozak KN and Harbour JW: PRAME as an independent biomarker for metastasis in uveal melanoma. Clin Cancer Res. 22:1234–1242. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Field MG, Durante MA, Decatur CL, Tarlan B, Oelschlager KM, Stone JF, Kuznetsov J, Bowcock AM, Kurtenbach S and Harbour JW: Epigenetic reprogramming and aberrant expression of PRAME are associated with increased metastatic risk in Class 1 and Class 2 uveal melanomas. Oncotarget. 7:59209–59219. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Brown JS, Flitcroft DI, Ying GS, Francis EL, Schmid GF, Quinn GE and Stone RA: In vivo human choroidal thickness measurements: Evidence for diurnal fluctuations. Invest Ophthalmol Vis Sci. 50:5–12. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Wicks NL, Chan JW, Najera JA, Ciriello JM and Oancea E: UVA phototransduction drives early melanin synthesis in human melanocytes. Curr Biol. 21:1906–1911. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Szuścik I, Romanowska-Dixon B, Jakubowska B and Orlowska-Heitzman J: Uveal melanoma in patients with ocular or oculodermal melanocytosis. Klin Oczna. 110:380–383. 2008.(In Polish). PubMed/NCBI

39 

Kuzirian MS and Paradis S: Emerging themes in GABAergic synapse development. Prog Neurobiol. 95:68–87. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Zieger E, Lacalli TC, Pestarino M, Schubert M and Candiani S: The origin of dopaminergic systems in chordate brains: Insights from amphioxus. Int J Dev Biol. 61:749–761. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Vicente-Rodríguez M, Rojo Gonzalez L, Gramage E, Fernández-Calle R, Chen Y, Pérez-García C, Ferrer-Alcón M, Uribarri M, Bailey A and Herradón G: Pleiotrophin overexpression regulates amphetamine-induced reward and striatal dopaminergic denervation without changing the expression of dopamine D1 and D2 receptors: Implications for neuroinflammation. Eur Neuropsychopharmacol. 26:1794–1805. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Baig AM: DARK side of amphetamine and analogues: pharmacology, syndromic manifestation, and management of amphetamine addiction. ACS Chem Neurosci. 9:2299–2303. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Ortells MO and Arias HR: Neuronal networks of nicotine addiction. Int J Biochem Cell Biol. 42:1931–1935. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 19 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yi Q and Yi Q: A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma. Mol Med Rep 19: 1294-1301, 2019
APA
Yi, Q., & Yi, Q. (2019). A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma. Molecular Medicine Reports, 19, 1294-1301. https://doi.org/10.3892/mmr.2018.9766
MLA
Yi, Q., Zou, W."A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma". Molecular Medicine Reports 19.2 (2019): 1294-1301.
Chicago
Yi, Q., Zou, W."A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma". Molecular Medicine Reports 19, no. 2 (2019): 1294-1301. https://doi.org/10.3892/mmr.2018.9766