Open Access

Profiling of prognostic alternative splicing in melanoma

  • Authors:
    • Fu‑Chao Ma
    • Rong‑Quan He
    • Peng Lin
    • Jin‑Cai Zhong
    • Jie Ma
    • Hong Yang
    • Xiao‑Hua Hu
    • Gang Chen
  • View Affiliations

  • Published online on: June 7, 2019     https://doi.org/10.3892/ol.2019.10453
  • Pages: 1081-1088
  • Copyright: © Ma et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alternative splicing can lead to the coding of proteins that act as promoters of cancer, which is associated with the progression of cancer. However, to the best of our knowledge, no systematic survival analysis of alternative splicing in melanoma has previously been reported. The present study conducted an in‑depth analysis of integrated alternative splicing events detected in 96 patients with melanoma using data obtained from The Cancer Genome Atlas. Prognostic models and an alternative splicing correlation network were built for patients with melanoma. A total of 41,446 mRNA splicing events were detected in 9,780 genes and 2,348 alternative splicing events were identified to be significantly associated with overall survival of patients with melanoma. Of all the events used in the prognostic model, the model with alternate terminator alternative splicing events exhibited the highest efficiency for evaluating the outcome of patients with melanoma, with an area under the curve of 0.902. The present study identified prognostic predictors for melanoma and revealed alternative splicing networks in melanoma that could indicate underlying mechanisms.

Introduction

Melanoma, the most aggressive type of skin cancer, develops from melanocytes and accounts for 4% of all cancer types diagnosed in adolescents worldwide (13). While the incidence rate of melanoma is lower than other types of skin cancer, it can grow rapidly and metastasize when it is not detected and treated early (46). Therefore, late-stage melanoma can be difficult to treat and fatal (7). The 5-year survival rate ranges between 15 and 60% depending on the stage of the melanoma (4,8).

It is understood that disordered gene expression serves an important role in the development of cancer (9). Gene expression profiling can provide evidence to determine the diagnosis and identify prognostic markers or novel therapeutic targets in cancer (10,11). However, a limited number of previous studies have investigated the role of gene expression by examining alternative splicing events (9,12,13).

Alternative splicing can lead to the coding of several types of proteins. During alternative splicing, specific exons of a gene can be included or excluded and the alternatively spliced mRNAs are then translated into proteins that exhibit different biological functions (1214). Alternative splicing regulates gene expression and serves a key role in this process (9). Defects in mRNA splicing can result in aberrant splicing, in addition to oncogenic processes (1517). A number of previous studies have demonstrated that splicing defects act as promoters of cancer (16,1820). Furthermore, different splicing factors may cause specific cancer-promoting isoforms.

It has been demonstrated that alternative splicing exhibits prognostic value in patients with lung (21), ovarian (22) and breast cancer (23), and glioblastoma (24). However, to the best of our knowledge, a systematic survival analysis of alternative splicing in melanoma has not been previously performed, and thus is urgently required. The aim of the present study was to determine prognostic alternative splicing events in melanoma using RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA).

Materials and methods

Alternative splicing data collection

Melanoma cohort RNA-seq data (25) were downloaded from TCGA data portal (tcga-data.nci.nih.gov/tcga). Data of 96 melanoma cases with clinicopathological information were obtained to explore the changes of alternative splicing events in association with the carcinogenesis and prognosis of melanoma. To analyze the alternative splicing profiles for each patient, a SpliceSeq tool (version 2.1) (26), which is a java application, was used in the melanoma cohort to evaluate the splicing patterns of mRNA. The percent spliced in (PSI) value was calculated to quantify alternative splicing events ranging between 0 and 1 in seven types of alternative splicing events, including exon skip (ES), alternate terminator (AT), mutually exclusive (ME) exons, retained intron (RI), alternate promoter (AP), alternate acceptor (AA) site and alternate donor (AD) site.

Survival analysis

The melanoma clinical data were downloaded from TCGA database. Only patients with an overall survival (OS) time >90 days were enrolled in the study. A total of 96 patients with melanoma were included in the present study. The patients were divided into two groups (high- and low-risk groups) according to the median cut-off value (0.078). Kaplan-Meier and Univariate Cox regression analyses were used to analyze associations between splicing factor genes and survival, and multivariate Cox regression was performed to remove non-independent predictors. To compare the efficiencies of each prediction model, receiver operator characteristic (ROC) curves were generated using the survival receiver ROC package (version 1.0.3; cran.r-project.org/web/packages/survivalROC/index.html) in R software (version 3.3.0; cran.r-project.org/bin/windows/base/old/3.3.0). To ascertain the predictive ability, the area under the curve (AUC) was calculated for each model.

Statistical analysis

The UpSet package (version 1.3.3; CRAN.R-project.org/package=UpSetR) in R software was used to quantitatively analyze the alternative splicing intersections for all seven types of alternative splicing events (27). The most significant alternative splicing events were selected to generate a gene network using the Reactome FI Cytoscape 3.6.0 plugin to search for the key hub genes of survival associated with the alternative splicing genes (28). Cytoscape 3.6.0 was then used to generate correlation plots. Spearman's rank correlation coefficient was used to analyze the correlation between the PSI values and the splicing factor gene expression to identify potential prognostic biomarkers for melanoma.

Results

Overview of alternative splicing events in TCGA melanoma cohort

An in-depth analysis of integrated alternative splicing events was conducted for 96 patients with melanoma with an OS time >90 days. In the melanoma cohort, 41,446 mRNA splicing events were detected in 9,780 genes, which consisted of 15,892 ESs in 6,160 genes, 8,267 ATs in 3,614 genes, 8,135 APs in 3,273 genes and 3,315 AAs in 2,350 genes, 2,968 ADs in 2,070 genes, 2,656 RIs in 1,780 genes and 177 MEs in 175 genes. The results suggested that several mRNA splicing events occur in one gene and ~38% of the alternative splicing events were ES events, followed by 19.9% AT events and 19.6% AP events (Fig. 1).

Survival-associated alternative splicing events in TCGA melanoma cohort

Every alternative splicing event in TCGA melanoma cohort was evaluated for its association with survival. The patients with melanoma were divided into two groups (high- and low-risk groups) based on the median PSI value. A total of 2,348 survival-associated alternative splicing events (data not shown) were detected in melanoma (P<0.05). The top ten most significant events in each type of alternative splicing event were selected. More than one alternative splicing event could occur in one gene, therefore, an UpSet plot was generated to visualize the intersecting sets. As demonstrated in Fig. 2, up to three types of splicing event were identified in one gene; however, the majority of the survival-associated alternative splicing events were from one gene and these were all associated with patient survival. Furthermore, a gene network was created by Cytoscape analysis. This revealed important cancer pathways in melanoma by the identification of the following hub genes: RNA polymerase II subunit I, epidermal growth factor receptor, nucleoporin 160, ribosomal protein S15A and ribosomal protein S3A (Fig. 3). Notably, numerous favorable prognostic factors were identified in the survival-associated alternative splicing events.

Prognostic predictors for patients with melanoma

The OS time of patients with melanoma ranged between 126 and 1,785 days. The top ten most significant events in the seven types of alternative splicing events were selected to determine the independent prognostic factors for patients with melanoma. Multivariate Cox regression analysis with a prognostic model construction was used to analyze the seven types of alternative splicing events separately, as demonstrated in Fig. 4. Kaplan-Meier curves of the prognostic predictors were generated for AA, AD, AP, AT, ES, ME and RI. The red line indicates the high-risk group and the blue line indicates the low-risk group. The seven predictors were integrated for all types of alternative splicing events in order to analyze the final prognostic model for patients with melanoma (Fig. 5). ROC curves were generated for the individual types of splicing event (Fig. 6) and for a combination of all types of splicing events (Fig. 7), and AUC analysis was used to evaluate the efficiency of the prognostic models. In the high-risk and low-risk groups, the median OS times were 472 and 1,354 days, respectively (P<0.0001; Fig. 5). Of all the models, the model with the AT alternative splicing events exhibited the highest efficiency for evaluating outcomes for patients with melanoma (Fig. 6D). The AUC value of the AT alternative splicing events was 0.902, followed by the AP model with an AUC value of 0.887 (Fig. 6C) and the RI model with an AUC value of 0.809 (Fig. 6G).

Alternative splicing correlation network in melanoma

A total of eight splicing factors, heterogeneous nuclear ribonucleoprotein L, serine and arginine rich splicing factor 9, Y-box binding protein 1, poly(rC) binding protein 2, heterogeneous nuclear ribonucleoprotein C (C1/C2), heterogeneous nuclear ribonucleoprotein A1, serine and arginine rich splicing factor 3 and serine and arginine rich splicing factor 4, were identified. As demonstrated in Fig. 8, these splicing factors, represented by green dots, were markedly associated with the OS time of the patients in the survival analysis. The expression levels and PSI values of these eight splicing factors identified from the most significant alternative splicing events were used to conduct the correlation analysis in the melanoma cohort based on the significant correlation results (P<0.05). As presented in Fig. 8, 50 survival-associated alternative splicing events were identified in the splicing correlation network, including 31 adverse alternative splicing events (red dots) and 19 favorable alternative splicing events (blue dots). The most favorable prognostic alternative splicing events (blue dots) were positively correlated (blue line) with the expression of the eight splicing factors (green dots), while the majority of the adverse prognosis alternative splicing events (red dots) were negatively correlated (red line) with the expression of the eight splicing factors.

Discussion

Previous studies have reported several aberrant alternative splicing events in melanoma (2831). Evidence has demonstrated that alternative splicing serves an important role in the occurrence and development of melanoma. For example, jumonji domain containing 6, arginine demethylase and lysine hydroxylase (JMJD6) regulates alternative splicing of p21 (RAC1) activated kinase 1, which is a key component of mitogen-activated protein kinase signaling; therefore, JMJD6 may promote carcinogenesis of melanoma (29). Alternative splicing of CD44 mediated by U2 small nuclear RNA auxiliary factor 2 promotes melanoma metastasis (30). However, thorough studies of alternative splicing may provide further information regarding the oncogenes and tumor suppressor genes of melanoma. Splicing factor 3b subunit 1 (SF3B1) is a key spliceosome component that was mutated in melanoma (30), chronic lymphocytic leukemia (32) and pancreatic cancer (33). Mutation of SF3B1 has been associated with good melanoma prognosis (31). A splicesome of human telomerase reverse transcriptase may control the activity of telomerase in melanoma. In the present study, 2,348 alternative splicing events were markedly associated with the OS time of patients with melanoma. The results of the present study are consistent with the findings reported in previous studies (29,30). These previous studies provide information regarding alternative splicing and splicing factors in melanoma, which may result in the identification of potential biomarkers and the development of therapeutic targets.

The development of high throughput sequencing technology has provided information regarding the features of alternative splicing (34,35). It is challenging to identify the splicesomes associated with cancer that determine the splicing patterns in the disease. Aberrant patterns of splicing leads to unlimited proliferation of cancer (36). The motility and invasion of cancer cells can be enhanced by serine and arginine-rich splicing factor 1, which modifies the expression of Ron-receptor tyrosine kinase (37). It was identified that vascular endothelial growth factor (VEGF) leads to angiogenesis in cancer. VEGF-A165, a splice variant of VEGF, is highly angiogenic; however, another splice variant of VEGF, VEGF-A165b, is downregulated in several types of cancer, such as colorectal cancer (38) and renal cell carcinoma (39). VEGF-A165b is associated with a poor prognosis of colorectal cancer (38). Although the role of splice variants is not completely understood, evidence indicates that they are strongly associated with specific types of cancer and exhibit potential prognostic value.

In the present study, alternative splicing features in 96 patients with melanoma were profiled using integrated survival analysis. A total of 41,446 alternative splicing events from 9,780 genes were detected and ~38% were ES events, followed by AT and AP events. ROC curves and Cox regression models were used to evaluate alternative splicing patterns as early biomarkers for melanoma occurrence or prognosis. Among the seven types of alternative splicing events, the AT events exhibited the highest efficiency for differentiating good or poor outcomes in patients with melanoma. The AUC in this model was 0.902, which is the highest value for all seven types of alternative splicing events, suggesting that it has the greatest potential for use as a prognostic marker for melanoma.

In addition, the present analysis highlighted the potential participation of splicing factors. The majority of the favorable prognosis alternative splicing events were positively correlated with the expression of splicing factors and the adverse prognosis alternative splicing events were negatively correlated with the expression of splicing factors. Expression of specific splicing factors may help determine if favorable or adverse alternative splicing events for prognosis require further validation by functional experiments. The present study investigated potential key splicing factors in melanoma and provided a background for understanding the role of splicesomes in melanoma.

In conclusion, the present study reported that alternative splicing events, which are associated with survival, may be used to generate a prognostic model for risk stratification in melanoma that could be applied in clinical practice. Furthermore, the present study provided novel insight into how pivotal splicing factors regulate aberrant alternative splicing. From the alternative splicing events and splicing factors that are associated with survival, several potential biomarkers or therapeutic targets were identified that require validation in future studies. Finally, a mechanism for deciphering alternative splicing events in tumorigenesis in melanoma was proposed.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets analyzed during the study are available from the corresponding author upon reasonable request.

Authors' contributions

GC and XHH designed the study. FCM, RQH, PL, JCZ, JM and HY conducted the experiments and analyzed the data. FM wrote the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

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 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Cui L, Li Y, Lv X, Li J, Wang X, Lei Z and Li X: Expression of MicroRNA-301a and its functional roles in malignant melanoma. Cell Physiol Biochem. 40:230–244. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Mahgoub T, Eustace AJ, Collins DM, Walsh N, O'Donovan N and Crown J: Kinase inhibitor screening identifies CDK4 as a potential therapeutic target for melanoma. Int J Oncol. 47:900–908. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Finn L, Markovic SN and Joseph RW: Therapy for metastatic melanoma: The past, present, and future. BMC Med. 10:232012. View Article : Google Scholar : PubMed/NCBI

5 

Carpinteiro A, Beckmann N, Seitz A, Hessler G, Wilker B, Soddemann M, Helfrich I, Edelmann B, Gulbins E and Becker KA: Role of acid sphingomyelinase-induced signaling in melanoma cells for hematogenous tumor metastasis. Cell Physiol Biochem. 38:1–14. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Pagani E, Ruffini F, Antonini Cappellini GC, Scoppola A, Fortes C, Marchetti P, Graziani G, D'Atri S and Lacal PM: Placenta growth factor and neuropilin-1 collaborate in promoting melanoma aggressiveness. Int J Oncol. 48:1581–1589. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Houghton AN and Polsky D: Focus on melanoma. Cancer Cell. 2:275–278. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Caporali S, Alvino E, Lacal PM, Levati L, Giurato G, Memoli D, Caprini E, Antonini Cappellini GC and D'Atri S: Targeting the PI3K/AKT/mTOR pathway overcomes the stimulating effect of dabrafenib on the invasive behavior of melanoma cells with acquired resistance to the BRAF inhibitor. Int J Oncol. 49:1164–1174. 2016. View Article : Google Scholar : PubMed/NCBI

9 

David CJ, Chen M, Assanah M, Canoll P and Manley JL: HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 463:364–368. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Zhang J, Shao J, Zhu L, Zhao R, Xing J, Wang J, Guo X, Tu S, Han B and Yu K: Molecular profiling identifies prognostic markers of stage IA lung adenocarcinoma. Oncotarget. 8:74846–74855. 2017.PubMed/NCBI

11 

Alaterre E, Raimbault S, Goldschmidt H, Bouhya S, Requirand G, Robert N, Boireau S, Seckinger, Hose D, Klein B and Moreaux J: CD24, CD27, CD36 and CD302 gene expression for outcome prediction in patients with multiple myeloma. Oncotarget. 8:98931–98944. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Kelemen O, Convertini P, Zhang Z, Wen Y, Shen M, Falaleeva M and Stamm S: Function of alternative splicing. Gene. 344:1–20. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Iqbal SM, Andavan GS and Lemmens-Gruber R: Differential modulation of fast inactivation in cardiac sodium channel splice variants by Fyn tyrosine kinase. Cell Physiol Biochem. 37:825–837. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Avin BA, Umbricht CB and Zeiger MA: Human telomerase reverse transcriptase regulation by DNA methylation, transcription factor binding and alternative splicing (Review). Int J Oncol. 49:2199–2205. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Bonomi S, Gallo S, Catillo M, Pignataro D, Biamonti G and Ghigna C: Oncogenic alternative splicing switches: Role in cancer progression and prospects for therapy. Int J Cell Biol. 2013:9620382013. View Article : Google Scholar : PubMed/NCBI

16 

Fackenthal JD and Godley LA: Aberrant RNA splicing and its functional consequences in cancer cells. Dis Model Mech. 1:37–42. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Xu Q, Gao J and Li Z: Identification of a novel alternative splicing transcript variant of the suppressor of fused: Relationship with lymph node metastasis in pancreatic ductal adenocarcinoma. Int J Oncol. 49:2611–2619. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Izaguirre DI, Zhu W, Hai T, Cheung HC, Krahe R and Cote GJ: PTBP1-dependent regulation of USP5 alternative RNA splicing plays a role in glioblastoma tumorigenesis. Mol Carcinog. 51:895–906. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Dhir A and Buratti E: Alternative splicing: Role of pseudoexons in human disease and potential therapeutic strategies. FEBS J. 277:841–855. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Karni R, de Stanchina E, Lowe SW, Sinha R, Mu D and Krainer AR: The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nat Struct Mol Biol. 14:185–193. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Li Y, Sun N, Lu Z, Sun S, Huang J, Chen Z and He J: Prognostic alternative mRNA splicing signature in non-small cell lung cancer. Cancer Lett. 393:40–51. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Zhu J, Chen Z and Yong L: Systematic profiling of alternative splicing signature reveals prognostic predictor for ovarian cancer. Gynecol Oncol. 148:368–374. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Suo C, Hrydziuszko O, Lee D, Pramana S, Saputra D, Joshi H, Calza S and Pawitan Y: Integration of somatic mutation, expression and functional data reveals potential driver genes predictive of breast cancer survival. Bioinformatics. 31:2607–2613. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Pal S, Bi Y, Macyszyn L, Showe LC, O'Rourke DM and Davuluri RV: Isoform-level gene signature improves prognostic stratification and accurately classifies glioblastoma subtypes. Nucleic Acids Res. 42:e642014. View Article : Google Scholar : PubMed/NCBI

25 

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. 32:204–220.e15. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Ryan MC, Cleland J, Kim R, Wong WC and Weinstein JN: SpliceSeq: A resource for analysis and visualization of RNA-Seq data on alternative splicing and its functional impacts. Bioinformatics. 28:2385–2387. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Lex A, Gehlenborg N, Strobelt H, Vuillemot R and Pfister H: UpSet: Visualization of intersecting sets. IEEE Trans Vis Comput Graph. 20:1983–1992. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Wu G, Feng X and Stein L: A human functional protein interaction network and its application to cancer data analysis. Genome Biol. 11:R532010. View Article : Google Scholar : PubMed/NCBI

29 

Liu X, Si W, Liu X, He L, Ren J, Yang Z, Yang J, Li W, Liu S, Pei F, et al: JMJD6 promotes melanoma carcinogenesis through regulation of the alternative splicing of PAK1, a key MAPK signaling component. Mol Cancer. 16:1752017. View Article : Google Scholar : PubMed/NCBI

30 

Zhang P, Feng S, Liu G, Wang H, Fu A, Zhu H, Ren Q, Wang B, Xu X, Bai H and Dong C: CD82 suppresses CD44 alternative splicing-dependent melanoma metastasis by mediating U2AF2 ubiquitination and degradation. Oncogene. 35:5056–5069. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Furney SJ, Pedersen M, Gentien D, Dumont AG, Rapinat A, Desjardins L, Turajlic S, Piperno-Neumann S, de la Grange P, Roman-Roman S, et al: SF3B1 mutations are associated with alternative splicing in uveal melanoma. Cancer Discov. 3:1122–1129. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Quesada V, Conde L, Villamor N, Ordóñez GR, Jares P, Bassaganyas L, Ramsay AJ, Beà S, Pinyol M, Martínez-Trillos A, et al: Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat Genet. 44:47–52. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, et al: Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 491:399–405. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Pan Q, Shai O, Lee LJ, Frey BJ and Blencowe BJ: Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 40:1413–1415. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Baty F, Klingbiel D, Zappa F and Brutsche M: High-throughput alternative splicing detection using dually constrained correspondence analysis (DCCA). J Biomed Inform. 58:175–185. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Oltean S and Bates DO: Hallmarks of alternative splicing in cancer. Oncogene. 33:5311–5318. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Das S and Krainer AR: Emerging functions of SRSF1, splicing factor and oncoprotein, in RNA metabolism and cancer. Mol Cancer Res. 12:1195–1204. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Díaz R, Peña C, Silva J, Lorenzo Y, García V, García JM, Sánchez A, Espinosa P, Yuste R, Bonilla F and Domínguez G: p73 isoforms affect VEGF, VEGF165b and PEDF expression in human colorectal tumours. VEGF165b downregulation as a marker for poor prognosis. Int J Cancer. 123:1060–1067. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Bates DO, Cui TG, Doughty JM, Winkler M, Sugiono M, Shields JD, Peat D, Gillatt D and Harper SJ: VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, is down-regulated in renal cell carcinoma. Cancer Res. 62:4123–4131. 2002.PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ma FC, He RQ, Lin P, Zhong JC, Ma J, Yang H, Hu XH and Chen G: Profiling of prognostic alternative splicing in melanoma. Oncol Lett 18: 1081-1088, 2019
APA
Ma, F., He, R., Lin, P., Zhong, J., Ma, J., Yang, H. ... Chen, G. (2019). Profiling of prognostic alternative splicing in melanoma. Oncology Letters, 18, 1081-1088. https://doi.org/10.3892/ol.2019.10453
MLA
Ma, F., He, R., Lin, P., Zhong, J., Ma, J., Yang, H., Hu, X., Chen, G."Profiling of prognostic alternative splicing in melanoma". Oncology Letters 18.2 (2019): 1081-1088.
Chicago
Ma, F., He, R., Lin, P., Zhong, J., Ma, J., Yang, H., Hu, X., Chen, G."Profiling of prognostic alternative splicing in melanoma". Oncology Letters 18, no. 2 (2019): 1081-1088. https://doi.org/10.3892/ol.2019.10453