Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Alternative splicing events in tumor immune infiltration in renal clear cell carcinomas

Abstract

Alternative splicing (AS) is a gene regulatory mechanism that drives protein diversity and dysregulation of AS plays a significant role in tumorigenesis. This study aimed to develop a prognostic signature based on AS and elucidate the role in tumor immune microenvironment (TIME) in clear cell renal cell carcinoma (ccRCC). The prognosis-related AS events were analyzed by univariate Cox regression analysis. Gene set enrichment analyses (GSEA) were performed for functional annotation. Prognostic signatures were identified and validated using univariate and multivariate Cox regression, LASSO regression, Kaplan–Meier survival analysis, and proportional hazards model. The context of TIME in ccRCC was also analyzed. Gene and protein expression data of C4orf19 were obtained from ONCOMINE website and Human Protein Altas. Splicing factors (SFs) regulatory networks were visualized. 4431 survival-related AS events in ccRCC were screened. Based on splicing subtypes, eight AS prognostic signatures were constructed. A nomogram with good prognostic prediction was generated. Furthermore, the prognostic signatures were significantly correlated with TIME diversity and immune checkpoint inhibitor (ICI)-related genes. C4orf19 was the only gene whose expression levels were downregulated among the prognostic AS-related genes, which is considered as a promising prognostic factor in ccRCC. Potential functions of SFs were determined by splicing regulatory networks. In our study, AS patterns of novel indicators for prognostic prediction of ccRCC were explored. The AS-SF networks provide information of regulatory mechanisms. Players of AS events related to TIME were investigated, which contribute to prognosis monitoring of ccRCC.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overall study design.
Fig. 2: Different AS types in ccRCC.
Fig. 3: The survival-related AS events.
Fig. 4: Confirmation of ALL AS-based prognostic signature.
Fig. 5: Correlation of ALL prognostic signature with clinical features and construction of AS-clinicopathological nomogram.
Fig. 6: Correlation between infiltrating immune cells and ALL AS-based prognostic signature.
Fig. 7: Association between ALL AS-based prognostic signature and key ICI genes.
Fig. 8: The clinical significance of C4orf19 in ccRCC.
Fig. 9: The role of C4orf19 in TIME features.
Fig. 10: The regulatory network between SFs and survival-related AS events.

Similar content being viewed by others

Data availability

Publicly available datasets were analyzed in this study.

References

  1. Motzer RJ, Chang SS, Jonasch E, Choueiri TK, Hancock SL, Lin DW, et al. Kidney cancer, version 3.2015. Pract Guide. 2015;13:151–9.

    Google Scholar 

  2. Gupta K, Miller JD, Li JZ, Russell MW, Charbonneau C. Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): a literature review. Cancer Treat Rev. 2008;34:193–205.

    Article  Google Scholar 

  3. Zuo Y, Zhang L, Tang W, Tang W. Identification of prognosis-related alternative splicing events in kidney renal clear cell carcinoma. J Cell Mol Med. 2019;23:7762–72.

    Article  CAS  Google Scholar 

  4. Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer–preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol. 2010;37:430–9.

    Article  CAS  Google Scholar 

  5. Kruger S, Ilmer M, Kobold S, Cadilha BL, Endres S, Ormanns S, et al. Advances in cancer immunotherapy 2019 - latest trends. J Exp Clin Cancer Res. 2019;38:268.

    Article  Google Scholar 

  6. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168:613–28.

    Article  CAS  Google Scholar 

  7. Montes M, Sanford BL, Comiskey DF, Chandler DS. RNA splicing and disease: animal models to therapies. Trends Genet. 2019;35:68–87.

    Article  CAS  Google Scholar 

  8. Frankiw L, Baltimore D, Li G. Alternative mRNA splicing in cancer immunotherapy. Nat Rev Immunol. 2019;19:675–87.

    Article  CAS  Google Scholar 

  9. Wu HY, Peng ZG, He RQ, Luo B, Ma J, Hu XH, et al. Prognostic index of aberrant mRNA splicing profiling acts as a predictive indicator for hepatocellular carcinoma based on TCGA SpliceSeq data. Int J Oncol. 2019;55:425–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Seiler M, Peng S, Agrawal AA, Palacino J, Teng T, Zhu P, et al. Somatic mutational landscape of splicing factor genes and their functional consequences across 33 cancer types. Cell Rep. 2018;23:282–96. e4

    Article  CAS  Google Scholar 

  11. Kouyama Y, Masuda T, Fujii A, Ogawa Y, Sato K, Tobo T, et al. Oncogenic splicing abnormalities induced by DEAD-Box Helicase 56 amplification in colorectal cancer. Cancer Sci. 2019;110:3132–44.

    Article  CAS  Google Scholar 

  12. Lee SC, Abdel-Wahab O. Therapeutic targeting of splicing in cancer. Nat Med. 2016;22:976–86.

    Article  CAS  Google Scholar 

  13. Zhou M, Diao Z, Cheng L, Sun J. Construction and analysis of dysregulated lncRNA-associated ceRNA network identified novel lncRNA biomarkers for early diagnosis of human pancreatic cancer. Oncotarget. 2016;7:56383–94.

    Article  Google Scholar 

  14. Wang C, Zheng M, Wang S, Nie X, Guo Q, Gao L, et al. Whole genome analysis and prognostic model construction based on alternative splicing events in endometrial cancer. Biomed Res Int. 2019;2019:2686875.

    PubMed  PubMed Central  Google Scholar 

  15. Meng T, Huang R, Zeng Z, Huang Z, Yin H, Jiao C, et al. Identification of prognostic and metastatic alternative splicing signatures in kidney renal clear cell carcinoma. Front Bioeng Biotechnol. 2019;7:270.

    Article  Google Scholar 

  16. Xiao L, Zou G, Cheng R, Wang P, Ma K, Cao H, et al. Alternative splicing associated with cancer stemness in kidney renal clear cell carcinoma. BMC Cancer. 2021;21:703.

    Article  CAS  Google Scholar 

  17. Ryan MC, Cleland J, Kim R, Wong WC, Weinstein JN. SpliceSeq: a resource for analysis and visualization of RNA-Seq data on alternative splicing and its functional impacts. Bioinformatics. 2012;28:2385–7.

    Article  CAS  Google Scholar 

  18. Blanche P, Dartigues JF, Jacqmin-Gadda H. Estimating and comparing time-dependent areas under receiver operating characteristic curves for censored event times with competing risks. Stat Med. 2013;32:5381–97.

    Article  Google Scholar 

  19. Goodman A, Patel SP, Kurzrock R. PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nat Rev Clin Oncol. 2017;14:203–20.

    Article  CAS  Google Scholar 

  20. Xu Q, Xu H, Deng R, Li N, Mu R, Qi Z, et al. Immunological significance of prognostic alternative splicing signature in hepatocellular carcinoma. Cancer Cell Int. 2021;21:190.

    Article  CAS  Google Scholar 

  21. Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin Cancer Res. 2017;23:124–36.

    Article  CAS  Google Scholar 

  22. Zhai L, Ladomersky E, Lenzen A, Nguyen B, Patel R, Lauing KL, et al. IDO1 in cancer: a Gemini of immune checkpoints. Cell Mol Immunol. 2018;15:447–57.

    Article  CAS  Google Scholar 

  23. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: response evaluation and biomarker development. Nat Rev Clin Oncol. 2017;14:655–68.

    Article  CAS  Google Scholar 

  24. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33.

    Article  Google Scholar 

  25. Heidegger I, Pircher A, Pichler R. Targeting the tumor microenvironment in renal cell cancer biology and therapy. Front Oncol. 2019;9:490.

    Article  Google Scholar 

  26. Finke JH, Rayman P, Edinger M, Tubbs RR, Stanley J, Klein E, et al. Characterization of a human renal cell carcinoma specific cytotoxic CD8+ T cell line. J Immunother. 1992;11:1–11.

    Article  CAS  Google Scholar 

  27. Fyfe BG, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. Clin Trial. 1995;13:688–96.

    CAS  Google Scholar 

  28. Figlin RA, Belldegrun A, Moldawer N, Zeffren J, deKernion J. Concomitant administration of recombinant human interleukin-2 and recombinant interferon alfa-2A: an active outpatient regimen in metastatic renal cell carcinoma. Clin Trial. 1992;10:414–21.

    CAS  Google Scholar 

  29. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Pr Guide. 2019;30:706–20.

    CAS  Google Scholar 

  30. Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, et al. The role of immunotherapy in solid tumors: report from the Campania Society of Oncology Immunotherapy (SCITO) meeting, Naples 2014. J Transl Med. 2014;12:291.

    Article  Google Scholar 

  31. Prieto J, Melero I, Sangro B. Immunological landscape and immunotherapy of hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2015;12:681–700.

    Article  CAS  Google Scholar 

  32. Hu J, Chen Z, Bao L, Zhou L, Hou Y, Liu L, et al. Single-cell transcriptome analysis reveals intratumoral heterogeneity in ccRCC, which results in different clinical outcomes. Mol Ther. 2020;28:1658–72.

    Article  CAS  Google Scholar 

  33. Nilsen TW, Graveley BR. Expansion of the eukaryotic proteome by alternative splicing. Nature. 2010;463:457–63.

    Article  CAS  Google Scholar 

  34. Climente-Gonzalez H, Porta-Pardo E, Godzik A, Eyras E. The functional impact of alternative splicing in cancer. Cell Rep. 2017;20:2215–26.

    Article  CAS  Google Scholar 

  35. Chevrier S, Levine JH, Zanotelli VRT, Silina K, Schulz D, Bacac M, et al. An immune atlas of clear cell renal cell carcinoma. Cell. 2017;169:736–49. e18

    Article  CAS  Google Scholar 

  36. Lang ZQ, Wu YQ, Pan XB, Qu GM, Zhang TG. The identification of multifocal breast cancer-associated long non-coding RNAs. Eur Rev Med Pharm Sci. 2017;21:5648–54.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

ZW, LP, KL, and PZ designed and supervised the study. ZW, LZ, LL, YS, and LP analyzed the data and wrote the original draft. LP, GG, and JS edited the draft. All the authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Ling Peng or Zhentao Yu.

Ethics declarations

Competing interests

JS’ conflicts can be found at https://www.nature.com/onc/editors. GG is Editor in Chief in Cancer Gene Therapy and the Founder and Chief Scientific Advisor of Stingray Bio. None are relevant here. Other authors do not declare conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Z., Zhu, L., Li, K. et al. Alternative splicing events in tumor immune infiltration in renal clear cell carcinomas. Cancer Gene Ther 29, 1418–1428 (2022). https://doi.org/10.1038/s41417-022-00426-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41417-022-00426-9

Search

Quick links