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.

  • Letter
  • Published:

Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins

An Erratum to this article was published on 07 June 2016

This article has been updated

Abstract

Mutations in genes encoding splicing factors (which we refer to as spliceosomal genes) are commonly found in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML)1,2,3. These mutations recurrently affect specific amino acid residues, leading to perturbed normal splice site and exon recognition4,5,6. Spliceosomal gene mutations are always heterozygous and rarely occur together with one another, suggesting that cells may tolerate only a partial deviation from normal splicing activity. To test this hypothesis, we engineered mice to express a mutated allele of serine/arginine-rich splicing factor 2 (Srsf2P95H)—which commonly occurs in individuals with MDS and AML—in an inducible, hemizygous manner in hematopoietic cells. These mice rapidly succumbed to fatal bone marrow failure, demonstrating that Srsf2-mutated cells depend on the wild-type Srsf2 allele for survival. In the context of leukemia, treatment with the spliceosome inhibitor E7107 (refs. 7,8) resulted in substantial reductions in leukemic burden, specifically in isogenic mouse leukemias and patient-derived xenograft AMLs carrying spliceosomal mutations. Whereas E7107 treatment of mice resulted in widespread intron retention and cassette exon skipping in leukemic cells regardless of Srsf2 genotype, the magnitude of splicing inhibition following E7107 treatment was greater in Srsf2-mutated than in Srsf2-wild-type leukemia, consistent with the differential effect of E7107 on survival. Collectively, these data provide genetic and pharmacologic evidence that leukemias with spliceosomal gene mutations are preferentially susceptible to additional splicing perturbations in vivo as compared to leukemias without such mutations. Modulation of spliceosome function may thus provide a new therapeutic avenue in genetically defined subsets of individuals with MDS or AML.

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

Figure 1: Spliceosome-mutant cells require the wild-type Srsf2 allele for survival.
Figure 2: SRSF2-mutated myeloid leukemias are preferentially sensitive to pharmacologic modulation of splicing catalysis.
Figure 3: Splicing and gene expression changes in Srsf2-WT or Srsf2-mutated leukemia cells that were treated with E7107.
Figure 4: Preferential sensitivity of primary human leukemias to pharmacologic modulation of splicing in vivo with E7107.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Sequence Read Archive

Change history

  • 11 May 2016

    In the version of this article initially published online, the graphs in Figure 3b–d were laid out incorrectly and were not consistent with the figure legend and text. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Yoshida, K. et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 478, 64–69 (2011).

    CAS  PubMed  Google Scholar 

  2. Papaemmanuil, E. et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N. Engl. J. Med. 365, 1384–1395 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Graubert, T.A. et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat. Genet. 44, 53–57 (2012).

    CAS  Google Scholar 

  4. Shirai, C.L. et al. Mutant U2AF1 expression alters hematopoiesis and pre-mRNA splicing in vivo. Cancer Cell 27, 631–643 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kim, E. et al. SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition. Cancer Cell 27, 617–630 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ilagan, J.O. et al. U2AF1 mutations alter splice site recognition in hematological malignancies. Genome Res. 25, 14–26 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Folco, E.G., Coil, K.E. & Reed, R. The antitumor drug E7107 reveals an essential role for SF3b in remodeling U2 snRNP to expose the branch point-binding region. Genes Dev. 25, 440–444 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Kotake, Y. et al. Splicing factor SF3b as a target of the antitumor natural product pladienolide. Nat. Chem. Biol. 3, 570–575 (2007).

    CAS  PubMed  Google Scholar 

  9. Lindsley, R.C. et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood 125, 1367–1376 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Genovese, G., Jaiswal, S., Ebert, B.L. & McCarroll, S.A. Clonal hematopoiesis and blood-cancer risk. N. Engl. J. Med. 372, 1071–1072 (2015).

    CAS  PubMed  Google Scholar 

  11. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    PubMed  PubMed Central  Google Scholar 

  13. Katz, Y., Wang, E.T., Airoldi, E.M. & Burge, C.B. Analysis and design of RNA sequencing experiments for identifying isoform regulation. Nat. Methods 7, 1009–1015 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Wagenmakers, E.J., Lodewyckx, T., Kuriyal, H. & Grasman, R. Bayesian hypothesis testing for psychologists: a tutorial on the Savage–Dickey method. Cognit. Psychol. 60, 158–189 (2010).

    PubMed  Google Scholar 

  15. Zhang, J. et al. Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc. Natl. Acad. Sci. USA 112, E4726–E4734 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lavallée, V.P. et al. The transcriptomic landscape and directed chemical interrogation of MLL-rearranged acute myeloid leukemias. Nat. Genet. 47, 1030–1037 (2015).

    PubMed  Google Scholar 

  17. Daigle, S.R. et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Bernt, K.M. et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell 20, 66–78 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Yokoi, A. et al. Biological validation that SF3b is a target of the antitumor macrolide pladienolide. FEBS J. 278, 4870–4880 (2011).

    CAS  PubMed  Google Scholar 

  20. Papaemmanuil, E. et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 122, 3616–3627, quiz 3699 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Bejar, R. et al. Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes. J. Clin. Oncol. 30, 3376–3382 (2012).

    PubMed  PubMed Central  Google Scholar 

  22. Zhang, S.J. et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood 119, 4480–4485 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Vannucchi, A.M. et al. Mutations and prognosis in primary myelofibrosis. Leukemia 27, 1861–1869 (2013).

    CAS  PubMed  Google Scholar 

  24. Effenberger, K.A., Urabe, V.K., Prichard, B.E., Ghosh, A.K. & Jurica, M.S. Interchangeable SF3B1 inhibitors interfere with pre-mRNA splicing at multiple stages. RNA 22, 350–359 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bonnal, S., Vigevani, L. & Valcárcel, J. The spliceosome as a target of novel antitumor drugs. Nat. Rev. Drug Discov. 11, 847–859 (2012).

    CAS  PubMed  Google Scholar 

  26. Eskens, F.A. et al. Phase 1 pharmacokinetic and pharmacodynamic study of the first-in-class spliceosome inhibitor E7107 in patients with advanced solid tumors. Clin. Cancer Res. 19, 6296–6304 (2013).

    CAS  PubMed  Google Scholar 

  27. Hong, D.S. et al. A phase 1, open-label, single-arm, dose-escalation study of E7107, a precursor messenger ribonucleic acid (pre-mRNA) splicesome inhibitor, administered intravenously on days 1 and 8 every 21 days to patients with solid tumors. Invest. New Drugs 32, 436–444 (2014).

    CAS  PubMed  Google Scholar 

  28. Katz, Y. et al. Quantitative visualization of alternative exon expression from RNA-seq data. Bioinformatics 31, 2400–2402 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Thorvaldsdóttir, H., Robinson, J.T. & Mesirov, J.P. Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    PubMed  Google Scholar 

  30. National Research Council. Guide for the Care and Use of Laboratory Animals 8th edn. (The National Academies Press, 2011).

  31. Wang, H.Y., Xu, X., Ding, J.H., Bermingham, J.R. Jr. & Fu, X.D. SC35 plays a role in T cell development and alternative splicing of CD45. Mol. Cell 7, 331–342 (2001).

    CAS  PubMed  Google Scholar 

  32. Georgiades, P. et al. Vav-Cre transgenic mice: a tool for mutagenesis in hematopoietic and endothelial lineages. Genesis 34, 251–256 (2002).

    CAS  PubMed  Google Scholar 

  33. Cheng, D.T. et al. Memorial Sloan Kettering–integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization-capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Meyer, L.R. et al. The UCSC Genome Browser database: extensions and updates 2013. Nucleic Acids Res. 41, D64–D69 (2013).

    CAS  PubMed  Google Scholar 

  35. Flicek, P. et al. Ensembl 2013. Nucleic Acids Res. 41, D48–D55 (2013).

    CAS  PubMed  Google Scholar 

  36. Hubert, C.G. et al. Genome-wide RNAi screens in human brain tumor isolates reveal a novel viability requirement for PHF5A. Genes Dev. 27, 1032–1045 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

  38. Li, B. & Dewey, C.N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  40. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Robinson, M.D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    PubMed  PubMed Central  Google Scholar 

  42. Young, M.D., Wakefield, M.J., Smyth, G.K. & Oshlack, A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome Biol. 11, R14 (2010).

    PubMed  PubMed Central  Google Scholar 

  43. Guberman, J.M. et al. BioMart central portal: an open database network for the biological community. Database 2011, bar041 (2011).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the Leukemia and Lymphoma Society (S.C.-W.L. and O.A.-W.), the US Department of Defense Breast Cancer Research Program grant W81XWH-14-1-0044 (H.D.), the US Department of Defense Bone Marrow Failure Research Program grants BM150092 (O.A.-W.) and W81XWH-12-1-0041 (R.K.B. and O.A.-W.), the Worldwide Cancer Research Fund (E.K.), the Fondation de France (J.-B.M.), the American Society of Hematology (B.H.D. and O.A.-W.), the Edward P. Evans Foundation (R.K.B. and O.A.-W.), the US National Institutes of Health (NIH)-NHLBI grant R01 HL128239 (R.K.B. and O.A.-W.), the NIH–NCI grant 1K08CA160647-01 (O.A.-W.), the Ellison Medical Foundation grant AG-NS-1030-13 (R.K.B.), the Damon Runyon Foundation (R.K.B. and O.A.-W.), the NIH–NIDDK grant R01 DK103854 (R.K.B.), the Starr Foundation grant I8-A8-075 (O.A.-W.), the Josie Robertson Investigator Program (O.A.-W.), the Mr. William H. Goodwin and Mrs. Alice Goodwin Commonwealth Foundation for Cancer Research (O.A.-W.), and the Experimental Therapeutics Center of MSKCC (O.A.-W.).

Author information

Authors and Affiliations

Authors

Contributions

S.C.-W.L., H.D., E.K., R.K.B., and O.A.-W. designed the study; S.C.-W.L., E.K., H.C., Y.R.C., J.-B.M., B.H.D., A.Y., Y.J.K., C.-W.C., A.P., J.T., X.W., A.K., S.A.A., and O.A.-W. performed experiments; H.D. and R.K.B. performed RNA-seq analysis; M.T., J.P., S.B., and P.G.S. provided E7107 and advice with drug-dosing experiments; and S.C.-W.L., H.D., E.K., C.L., R.K.B., and O.A.-W. prepared the manuscript with help from all co-authors.

Corresponding authors

Correspondence to Robert K Bradley or Omar Abdel-Wahab.

Ethics declarations

Competing interests

M.T., J.P., S.B., and P.G.S. are employees of H3 Biomedicine, Inc.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 2 and 4 (PDF 40706 kb)

Supplementary Table 1

Median normalized gene expression values across replicates in each of the murine mRNA sequencing experiments shown in Figures 1-3. (XLSX 2160 kb)

Supplementary Table 3

Median exon inclusion rates (PSI (percent spliced in) values (ψ)) for cassette exons across replicates in each of the murine mRNA sequencing experiments shown in Figures 1-3. (XLSX 1831 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, SW., Dvinge, H., Kim, E. et al. Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins. Nat Med 22, 672–678 (2016). https://doi.org/10.1038/nm.4097

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4097

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing