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:

Identification and in-silico characterization of splice-site variants from a large cardiogenetic national registry

Abstract

Splice-site variants in cardiac genes may predispose carriers to potentially lethal arrhythmias. To investigate, we screened 1315 probands and first-degree relatives enrolled in the Canadian Hearts in Rhythm Organization (HiRO) registry. 10% (134/1315) of patients in the HiRO registry carry variants within 10 base-pairs of the intron-exon boundary with 78% (104/134) otherwise genotype negative. These 134 probands were carriers of 57 unique variants. For each variant, American College of Medical Genetics and Genomics (ACMG) classification was revisited based on consensus between nine in silico tools. Due in part to the in silico algorithms, seven variants were reclassified from the original report, with the majority (6/7) downgraded. Our analyses predicted 53% (30/57) of variants to be likely/pathogenic. For the 57 variants, an average of 9 tools were able to score variants within splice sites, while 6.5 tools responded for variants outside these sites. With likely/pathogenic classification considered a positive outcome, the ACMG classification was used to calculate sensitivity/specificity of each tool. Among these, Combined Annotation Dependent Depletion (CADD) had good sensitivity (93%) and the highest response rate (131/134, 98%), dbscSNV was also sensitive (97%), and SpliceAI was the most specific (64%) tool. Splice variants remain an important consideration in gene elusive inherited arrhythmia syndromes. Screening for intronic variants, even when restricted to the ±10 positions as performed here may improve genetic testing yield. We compare 9 freely available in silico tools and provide recommendations regarding their predictive capabilities. Moreover, we highlight several novel cardiomyopathy-associated variants which merit further study.

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: RNA splicing schematic.
Fig. 2: Work-flow of utilized in silico tools.
Fig. 3: Position of identified splice-site variants.
Fig. 4: Predictive capability of in silico tools.

Similar content being viewed by others

Data availability

The data underlying this article are available within the text, appendices, and online supplementary material.

References

  1. Shapiro MB, Senapathy P. RNA splice junctions of different classes of eukaryotes: sequence statistics and functional implications in gene expression. Nucleic acids Res. 1987;15:7155–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zhang M. Statistical features of human exons and their flanking regions. Hum Mol Genet. 1998;7:919–32.

    Article  CAS  PubMed  Google Scholar 

  3. Walsh R, Thomson KL, Ware JS, Funke BH, Woodley J, McGuire KJ, et al. Reassessment of Mendelian gene pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference samples. Genet Med. 2017;19:192–203.

    Article  PubMed  Google Scholar 

  4. Bagnall RD, Ingles J, Dinger ME, Cowley MJ, Ross SB, Minoche AE, et al. Whole genome sequencing improves outcomes of genetic testing in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol. 2018;72:419–29.

    Article  PubMed  Google Scholar 

  5. Krawczak M, Reiss J, Cooper DN. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences. Hum Genet. 1992;90:41–54.

    Article  CAS  PubMed  Google Scholar 

  6. Veitia RA, Birchler JA. Dominance and gene dosage balance in health and disease: why levels matter! J Pathol: A J Pathological Soc Gt Br Irel. 2010;220:174–85.

    Article  CAS  Google Scholar 

  7. Moon H, Jang HN, Liu Y, Choi N, Oh J, Ha J, et al. Activation of cryptic 3’ splice-sites by SRSF2 contributes to cassette exon skipping. Cells. 2019;8:696.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jaganathan K, Panagiotopoulou SK, McRae JF, Darbandi SF, Knowles D, Li YI, et al. Predicting splicing from primary sequence with deep learning. Cell 2019;176:535–48. e24.

    Article  CAS  PubMed  Google Scholar 

  9. Jian X, Boerwinkle E, Liu X. In silico tools for splicing defect prediction: a survey from the viewpoint of end users. Genet Med. 2014;16:497–503.

    Article  CAS  PubMed  Google Scholar 

  10. Baralle D, Lucassen A, Buratti E. Missed threads. The impact of pre-mRNA splicing defects on clinical practice. EMBO Rep. 2009;10:810–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gasteiger E, Gattiker A, Hoogland C, Ivanyi I, Appel RD, Bairoch A. ExPASy: the proteomics server for in-depth protein knowledge and analysis. Nucleic acids Res. 2003;31:3784–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hubbard TJ, Aken BL, Ayling S, Ballester B, Beal K, Bragin E, et al. Ensembl 2009. Nucleic acids Res. 2009;37(suppl_1):D690–D7.

    Article  CAS  PubMed  Google Scholar 

  13. Reese MG, Eeckman FH, Kulp D, Haussler D. Improved splice site detection in Genie. J computational Biol. 1997;4:311–23.

    Article  CAS  Google Scholar 

  14. Schwarz JM, Rödelsperger C, Schuelke M, Seelow D. MutationTaster evaluates disease-causing potential of sequence alterations. Nat methods. 2010;7:575–6.

    Article  CAS  PubMed  Google Scholar 

  15. Kircher M, Witten DM, Jain P, O’Roak BJ, Cooper GM, Shendure J. A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet. 2014;46:310–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Quang D, Chen Y, Xie X. DANN: a deep learning approach for annotating the pathogenicity of genetic variants. Bioinformatics 2015;31:761–3.

    Article  CAS  PubMed  Google Scholar 

  17. Jian X, Boerwinkle E, Liu X. In silico prediction of splice-altering single nucleotide variants in the human genome. Nucleic acids Res. 2014;42:13534–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–23.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Davies B, Roberts JD, Tadros R, Green MS, Healey JS, Simpson CS, et al. The hearts in rhythm organization: a Canadian national cardiogenetics network. CJC open. 2020;2:652–62.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Aguilera MA, Meyer R, et al. VarSome: the human genomic variant search engine. Bioinformatics 2019;35:1978.

    Article  CAS  PubMed  Google Scholar 

  21. Karczewski KJ, Francioli LC, Tiao G, Cummings BB, Alföldi J, Wang Q, et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 2020;581:434–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu X, Jian X, Boerwinkle E. dbNSFP: a lightweight database of human nonsynonymous SNPs and their functional predictions. Hum Mutat. 2011;32:894–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073.

    Article  CAS  PubMed  Google Scholar 

  24. Adzhubei I, Jordan DM, Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen‐2. Curr Protoc in Hum Genet. 2013:7.20:1–7.

  25. Desmet F-O, Hamroun D, Lalande M, Collod-Béroud G, Claustres M, Béroud C. Human Splicing Finder: an online bioinformatics tool to predict splicing signals. Nucleic acids Res. 2009;37:e67–e.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Yeo G, Burge CB. Maximum entropy modeling of short sequence motifs with applications to RNA splicing signals. J computational Biol. 2004;11:377–94.

    Article  CAS  Google Scholar 

  27. Sun B, Yao J, Ni M, Wei J, Zhong X, Guo W, et al. Cardiac ryanodine receptor calcium release deficiency syndrome. Sci Transl Med. 2021;13:eaba7287.

    Article  CAS  PubMed  Google Scholar 

  28. Desmet F-O, Hamroun D, Collod-Béroud G, Claustres M, Béroud C. Bioinformatics identification of splice site signals and prediction of mutation effects. Global Research Network Publishers; 2010, pp. 1–14.

  29. Houdayer C, Caux‐Moncoutier V, Krieger S, Barrois M, Bonnet F, Bourdon V, et al. Guidelines for splicing analysis in molecular diagnosis derived from a set of 327 combined in silico/in vitro studies on BRCA1 and BRCA2 variants. Hum Mutat. 2012;33:1228–38.

    Article  CAS  PubMed  Google Scholar 

  30. Rowlands C, Thomas HB, Lord J, Wai HA, Arno G, Beaman G, et al. Comparison of in silico strategies to prioritize rare genomic variants impacting RNA splicing for the diagnosis of genomic disorders. Sci Rep. 2021;11:1–11.

    Article  Google Scholar 

  31. Anderson D, Lassmann T. A phenotype centric benchmark of variant prioritisation tools. NPJ Genom Med. 2018;3:1–9.

    Article  CAS  Google Scholar 

  32. Bhuiyan ZA, Momenah TS, Amin AS, Al-Khadra AS, Alders M, Wilde AA, et al. An intronic mutation leading to incomplete skipping of exon-2 in KCNQ1 rescues hearing in Jervell and Lange-Nielsen syndrome. Prog biophysics Mol Biol. 2008;98:319–27.

    Article  CAS  Google Scholar 

  33. Duggal P, Vesely MR, Wattanasirichaigoon D, Villafane J, Kaushik V, Beggs AH. Mutation of the gene for I sK associated with both Jervell and Lange-Nielsen and Romano-Ward forms of long-QT syndrome. Circulation 1998;97:142–6.

    Article  CAS  PubMed  Google Scholar 

  34. Campuzano O, Fernández-Falgueras A, Iglesias A, Brugada R Brugada Syndrome and PKP2: Evidences and uncertainties. Elsevier; 2016.

  35. Ben-Haim Y, Asimaki A, Behr ER Brugada syndrome and arrhythmogenic cardiomyopathy: overlapping disorders of the connexome? EP Europace. 2020.

  36. Mort M, Sterne-Weiler T, Li B, Ball EV, Cooper DN, Radivojac P, et al. MutPred Splice: machine learning-based prediction of exonic variants that disrupt splicing. Genome Biol. 2014;15:R19.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Leman R, Gaildrat P, Le Gac G, Ka C, Fichou Y, Audrezet MP, et al. Novel diagnostic tool for prediction of variant spliceogenicity derived from a set of 395 combined in silico/in vitro studies: an international collaborative effort. Nucleic acids Res. 2018;46:7913–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jagadeesh KA, Paggi JM, Ye JS, Stenson PD, Cooper DN. S-CAP extends pathogenicity prediction to genetic variants that affect RNA splicing. 2019;51:755–63.

  39. Cheng J, Nguyen TYD, Cygan KJ, Çelik MH, Fairbrother WG, Avsec Ž, et al. MMSplice: modular modeling improves the predictions of genetic variant effects on splicing. 2019;20:48.

  40. Danis D, Jacobsen JOB, Carmody LC, Gargano MA, McMurry JA, Hegde A, et al. Interpretable prioritization of splice variants in diagnostic next-generation sequencing. Am J Hum Genet. 2021;108:1564–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Rentzsch P, Schubach M, Shendure J, Kircher M. CADD-Splice—improving genome-wide variant effect prediction using deep learning-derived splice scores. Genome Med. 2021;13:31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Singer ES, Ingles J, Semsarian C, Bagnall RD. Key value of RNA analysis of MYBPC3 splice-site variants in hypertrophic cardiomyopathy. Circ Genom Precis Med. 2019;12:e002368.

    Article  CAS  PubMed  Google Scholar 

  43. Holliday M, Singer ES, Ross SB, Lim S, Lal S, Ingles J, et al. Transcriptome sequencing of patients with hypertrophic cardiomyopathy reveals novel splice-altering variants in MYBPC3. Circulation: Genomic and Precision Medicine. 2021.

  44. Heinig M, Adriaens ME, Schafer S, van Deutekom HW, Lodder EM, Ware JS, et al. Natural genetic variation of the cardiac transcriptome in non-diseased donors and patients with dilated cardiomyopathy. Genome Biol. 2017;18:1–21.

    Article  Google Scholar 

  45. Chaudhry F, Isherwood J, Bawa T, Patel D, Gurdziel K, Lanfear DE, et al. Single-cell RNA sequencing of the cardiovascular system: new looks for old diseases. Front cardiovascular Med. 2019;6:173.

    Article  CAS  Google Scholar 

  46. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10:57–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Black DL. Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem. 2003;72:291–336.

    Article  CAS  PubMed  Google Scholar 

  48. Ito K, Patel PN, Gorham JM, McDonough B, DePalma SR, Adler EE, et al. Identification of pathogenic gene mutations in LMNA and MYBPC3 that alter RNA splicing. Proc Natl Acad Sci 2017;114:7689–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wollnik B, Schroeder BC, Kubisch C, Esperer HD, Wieacker P, Jentsch TJ. Pathophysiological mechanisms of dominant and recessive KVLQT1 K+ channel mutations found in inherited cardiac arrhythmias. Hum Mol Genet. 1997;6:1943–9.

    Article  CAS  PubMed  Google Scholar 

  50. Ule J, Blencowe BJ. Alternative splicing regulatory networks: functions, mechanisms, and evolution. Mol cell. 2019;76:329–45.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by the Faculty of Medicine Summer Student Research Program at the University of British Columbia provided to KR. ZL was funded by the Michael Smith Foundation for Health Research and the Cardiology Academic Practice Plan at the University of British Columbia. The study was supported by the Heart in Rhythm Organization (Dr. Krahn, Principal Investigator) that receives support from the Canadian Institute of Health Research (RN380020 – 406814).

Author information

Authors and Affiliations

Authors

Contributions

Experimental design: KR, BD, RDB, ADK, ZWML. Data acquisition: KR, BD. Data analysis and interpretation: KR, BD, RDB, ADK, ZWML. Manuscript preparation: KR, BD, MC, DC, RDB, ADK, ZWML. Manuscript editting: KR, BD, MC, DC, JDR, RT, MSG, JSH, CSS, SS, CS, CM, PA, HD, RH, LA, RL, CS, AF, JA, SK, BM, WA, JC-T, JJ, MG, MT, RDB, ADK, ZWML.

Corresponding author

Correspondence to Zachary W. M. Laksman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Ethical approval

This project was conducted in compliance with the protocols and principles laid down in the Declaration of Helsinki and approved in full by the Providence Health/University of British Columbia ethics board (REB number H20-00129).

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

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rayani, K., Davies, B., Cheung, M. et al. Identification and in-silico characterization of splice-site variants from a large cardiogenetic national registry. Eur J Hum Genet 31, 512–520 (2023). https://doi.org/10.1038/s41431-022-01193-9

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41431-022-01193-9

This article is cited by

Search

Quick links