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:

U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies

Abstract

Spliceosome mutations are common in myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML), but the oncogenic changes due to these mutations have not been identified. Here a global analysis of exon usage in AML samples revealed distinct molecular subsets containing alternative spliced isoforms of inflammatory and immune genes. Interleukin-1 receptor-associated kinase 4 (IRAK4) was the dominant alternatively spliced isoform in MDS and AML and is characterized by a longer isoform that retains exon 4, which encodes IRAK4-long (IRAK4-L), a protein that assembles with the myddosome, results in maximal activation of nuclear factor kappa-light-chain-enhancer of B cells (NF-κB) and is essential for leukaemic cell function. Expression of IRAK4-L is mediated by mutant U2 small nuclear RNA auxiliary factor 1 (U2AF1) and is associated with oncogenic signalling in MDS and AML. Inhibition of IRAK4-L abrogates leukaemic growth, particularly in AML cells with higher expression of the IRAK4-L isoform. Collectively, mutations in U2AF1 induce expression of therapeutically targetable ‘active’ IRAK4 isoforms and provide a genetic link to activation of chronic innate immune signalling in MDS and 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

Fig. 1: Differential RNA isoform usage correlates with AML prognosis and oncogenic IRAK4 isoforms.
Fig. 2: IRAK4-L expression results in myddosome assembly and maximal activation of innate immune signalling.
Fig. 3: IRAK4-L is required for leukaemic cell function.
Fig. 4: IRAK4-L expression is associated with U2AF1 mutations in MDS and AML.
Fig. 5: U2AF1(S34F) induces expression of IRAK4-L and increased innate immune pathway activation.
Fig. 6: U2AF1(S34F) AML cells are sensitive to IRAK4 inhibitors.

Similar content being viewed by others

Data availability

RNA-sequencing data that support the findings of this study have been deposited in the Gene Expression Omnibus under accession codes GSE114922 (corresponding to data in Fig. 4b), GSE58831 (corresponding to data in Supplementary Fig. 4), dbGaP: phs000159.v8 (SRX1608652, SRX1608653, SRX1608605, SRX1608609, SRX1608616 and SRX1608620) (corresponding to data in Fig. 5a), and E-MTAB-1733 (corresponding to data in Fig. 1g). The human AML data were derived from the TCGA Research Network: http://cancergenome.nih.gov. Protein isoform-isoform interaction network was generated using human protein sequences (GRCh37/hg19) from the UCSC Genome Browser’s Table Browser (https://genome.ucsc.edu/cgi-bin/hgTables, accessed February 2009); protein-domain models from PFAM v28 (ftp://ftp.ebi.ac.uk/pub/databases/Pfam/releases/Pfam28.0/), CCD v3.16 (ftp://ftp.ncbi.nih.gov/pub/mmdb/cdd), and InterPro v50 (ftp://ftp.ebi.ac.uk/pub/databases/interpro/50.0/); protein–protein interactions from HPRD v9 (http://hprd.org/download); and domain–domain interactions from DOMINE v2.0 (https://manticore.niehs.nih.gov/cgi-bin/Domine?page = download) (corresponding to data in Fig. 2i). Source data for Figs. 26 and Supplementary Figs. 3, 4 and 6 have been provided as Supplementary Table 5. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Vitting-Seerup, K. & Sandelin, A. The landscape of isoform switches in human cancers. Mol. Cancer Res. 15, 1206–1220 (2017).

    Article  CAS  Google Scholar 

  2. Inoue, D., Bradley, R. K. & Abdel-Wahab, O. Spliceosomal gene mutations in myelodysplasia: molecular links to clonal abnormalities of hematopoiesis. Genes Dev. 30, 989–1001 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. 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).

    Article  CAS  Google Scholar 

  5. Saez, B., Walter, M. J. & Graubert, T. A. Splicing factor gene mutations in hematologic malignancies. Blood 129, 1260–1269 (2017).

    Article  CAS  Google Scholar 

  6. Graubert, T. A. et al. Recurrent mutations in the U2AF1 splicing factor in myelodysplastic syndromes. Nat. Genet. 44, 53–57 (2011).

    Article  Google Scholar 

  7. Hirsch, C. M. et al. Molecular features of early onset adult myelodysplastic syndrome. Haematologica 102, 1028–1034 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Pellagatti, A. & Boultwood, J. Splicing factor gene mutations in the myelodysplastic syndromes: impact on disease phenotype and therapeutic applications. Adv. Biol. Regul. 63, 59–70 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Haferlach, T. et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia 28, 241–247 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  13. Ley, T. J. et al. DNMT3A mutations in acute myeloid leukemia. N. Engl. J. Med. 363, 2424–2433 (2010).

    Article  CAS  Google Scholar 

  14. Barreyro, L., Chlon, T. M. & Starczynowski, D. T. Chronic immune response dysregulation in MDS pathogenesis. Blood 132, 1553–1560 (2018).

    Article  CAS  Google Scholar 

  15. Patra, M. C. & Choi, S. Recent progress in the molecular recognition and therapeutic importance of interleukin-1 receptor-associated kinase 4. Molecules 21, 1529 (2016).

    Article  Google Scholar 

  16. Yang, X. et al. Widespread expansion of protein interaction capabilities by alternative splicing. Cell 164, 805–817 (2016).

    Article  CAS  Google Scholar 

  17. Dossang, A. C. et al. The N-terminal loop of IRAK-4 death domain regulates ordered assembly of the Myddosome signalling scaffold. Sci. Rep. 6, 37267 (2016).

    Article  CAS  Google Scholar 

  18. Ferrao, R. et al. IRAK4 dimerization and trans-autophosphorylation are induced by Myddosome assembly. Mol. Cell 55, 891–903 (2014).

    Article  CAS  Google Scholar 

  19. Booher R. N., S.M., Xu G., Cheng H., Tuck D. P. Efficacy of the IRAK4 inhibitor CA-4948 in patient-derived xenograft models of diffuse large B cell lymphoma. In Proc. American Association for Cancer Research Annual Meeting 2017 (AACR, 2017).

  20. Gerstung, M. et al. Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes. Nat. Commun. 6, 5901 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Brooks, A. N. et al. A pan-cancer analysis of transcriptome changes associated with somatic mutations in U2AF1 reveals commonly altered splicing events. PLoS One 9, e87361 (2014).

    Article  Google Scholar 

  23. Fei, D. L. et al. Wild-Type U2AF1 antagonizes the splicing program characteristic of u2af1-mutant tumors and is required for cell survival. PLoS Genet. 12, e1006384 (2016).

    Article  Google Scholar 

  24. Yip, B. H. et al. The U2AF1 S34F mutation induces lineage-specific splicing alterations in myelodysplastic syndromes. J. Clin. Invest. 127, 2206–2221 (2017).

    Article  Google Scholar 

  25. Okeyo-Owuor, T. et al. U2AF1 mutations alter sequence specificity of pre-mRNA binding and splicing. Leukemia 29, 909–917 (2015).

    Article  CAS  Google Scholar 

  26. Shirai, C. L. et al. Mutant U2AF1-expressing cells are sensitive to pharmacological modulation of the spliceosome. Nat. Commun. 8, 14060 (2017).

    Article  CAS  Google Scholar 

  27. Powers, J. P. et al. Discovery and initial SAR of inhibitors of interleukin-1 receptor-associated kinase-4. Bioorg. Med. Chem. Lett. 16, 2842–2845 (2006).

    Article  CAS  Google Scholar 

  28. Maimon, A. et al. Mnk2 alternative splicing modulates the p38-MAPK pathway and impacts Ras-induced transformation. Cell Rep. 7, 501–513 (2014).

    Article  CAS  Google Scholar 

  29. Hofmann, W. K. et al. Characterization of gene expression of CD34+cells from normal and myelodysplastic bone marrow. Blood 100, 3553–3560 (2002).

    Article  CAS  Google Scholar 

  30. Pellagatti, A. et al. Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells. Leukemia 24, 756–764 (2010).

    Article  CAS  Google Scholar 

  31. Kristinsson, S. Y. et al. Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes. J. Clin. Oncol. 29, 2897–2903 (2011).

    Article  Google Scholar 

  32. Starczynowski, D. T. et al. Identification of miR-145 and miR-146a as mediators of the 5q– syndrome phenotype. Nat. Med. 16, 49–58 (2010).

    Article  CAS  Google Scholar 

  33. Varney, M. E. et al. Epistasis between TIFAB and miR-146a: neighboring genes in del(5q) myelodysplastic syndrome. Leukemia 31, 491–495 (2017).

    Article  CAS  Google Scholar 

  34. Varney, M. E. et al. Loss of Tifab, a del(5q) MDS gene, alters hematopoiesis through derepression of Toll-like receptor–TRAF6 signaling. J. Exp. Med. 212, 1967–1985 (2015).

    Article  CAS  Google Scholar 

  35. Rhyasen, G. W. et al. Targeting IRAK1 as a therapeutic approach for myelodysplastic syndrome. Cancer Cell 24, 90–104 (2013).

    Article  CAS  Google Scholar 

  36. Beverly, L. J. & Starczynowski, D. T. IRAK1: oncotarget in MDS and AML. Oncotarget 5, 1699–1700 (2014).

    Article  Google Scholar 

  37. Dussiau, C. et al. Targeting IRAK1 in T-cell acute lymphoblastic leukemia. Oncotarget 6, 18956–18965 (2015).

    Article  Google Scholar 

  38. Li, Z. et al. Inhibition of IRAK1/4 sensitizes T cell acute lymphoblastic leukemia to chemotherapies. J. Clin. Invest. 125, 1081–1097 (2015).

    Article  Google Scholar 

  39. Goh, J. Y. et al. Chromosome 1q21.3 amplification is a trackable biomarker and actionable target for breast cancer recurrence. Nat. Med. 23, 1319–1330 (2017).

    Article  CAS  Google Scholar 

  40. Adams, A. K. et al. IRAK1 is a novel DEK transcriptional target and is essential for head and neck cancer cell survival. Oncotarget 6, 43395–43407 (2015).

    PubMed  PubMed Central  Google Scholar 

  41. Wee, Z. N. et al. IRAK1 is a therapeutic target that drives breast cancer metastasis and resistance to paclitaxel. Nat. Commun. 6, 8746 (2015).

    Article  CAS  Google Scholar 

  42. Lee, S. C. et al. Synthetic lethal and convergent biological effects of cancer-associated spliceosomal gene mutations. Cancer Cell 34, 225–241 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  44. Makishima, H. et al. Mutations in the spliceosome machinery, a novel and ubiquitous pathway in leukemogenesis. Blood 119, 3203–3210 (2012).

    Article  CAS  Google Scholar 

  45. Ogawa, S. Splicing factor mutations in AML. Blood 123, 3216–3217 (2014).

    Article  CAS  Google Scholar 

  46. Kon, A. et al. Physiological Srsf2 P95H expression causes impaired hematopoietic stem cell functions and aberrant RNA splicing in mice. Blood 131, 621–635 (2018).

    Article  CAS  Google Scholar 

  47. Komeno, Y. et al. SRSF2 Is essential for hematopoiesis, and its myelodysplastic syndrome-related mutations dysregulate alternative pre-mRNA splicing. Mol. Cell Biol. 35, 3071–3082 (2015).

    Article  CAS  Google Scholar 

  48. Obeng, E. A. et al. Physiologic expression of Sf3b1 K700E causes impaired erythropoiesis, aberrant splicing, and sensitivity to therapeutic spliceosome modulation. Cancer Cell 30, 404–417 (2016).

    Article  CAS  Google Scholar 

  49. Mupo, A. et al. Hemopoietic-specific Sf3b1-K700E knock-in mice display the splicing defect seen in human MDS but develop anemia without ring sideroblasts. Leukemia 31, 720–727 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  51. Komurov, K., Dursun, S., Erdin, S. & Ram, P. T. NetWalker: a contextual network analysis tool for functional genomics. BMC Genomics 13, 282 (2012).

    Article  Google Scholar 

  52. Matsuoka, A. et al. Lenalidomide induces cell death in an MDS-derived cell line with deletion of chromosome 5q by inhibition of cytokinesis. Leukemia 24, 748–755 (2010).

    Article  CAS  Google Scholar 

  53. Rhyasen, G. W. et al. An MDS xenograft model utilizing a patient-derived cell line. Leukemia 28, 1142–1145 (2014).

    Article  CAS  Google Scholar 

  54. Sun, J. et al. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci. Signal. 9, aab2191 (2016).

    Article  Google Scholar 

  55. Komurov, K., White, M. A. & Ram, P. T. Use of data-biased random walks on graphs for the retrieval of context-specific networks from genomic data. PLoS Comput. Biol. 6, 1000889 (2010).

    Article  Google Scholar 

  56. Stoilov, P., Lin, C. H., Damoiseaux, R., Nikolic, J. & Black, D. L. A high-throughput screening strategy identifies cardiotonic steroids as alternative splicing modulators. Proc. Natl Acad. Sci. USA 105, 11218–11223 (2008).

    Article  CAS  Google Scholar 

  57. Fang, J. et al. Ubiquitination of the spliceosome auxiliary factor hnRNPA1 by TRAF6 links chronic innate immune signaling with hematopoietic defects and myelodysplasia. Nat. Immunol. 18, 236–245 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by Cincinnati Children’s Hospital Research Foundation, Leukemia Lymphoma Society, National Institutes of Health (R35HL135787, RO1DK102759, RO1DK113639), and Edward P. Evans Foundation grants to D.T.S. A.V. is supported by National Institutes of Health (R01HL139487 and R01DK103961), Leukemia and Lymphoma Society, and EvansMDS grants. A.P. and J.B. are supported by Bloodwise (UK, grant 13042). M.A.S. is supported by a National Institutes of Health Research Training and Career Development Grant (F31HL132420). I.D.C.F. is supported by the Intramural Research Program of NIAID. We also thank R. Booher (Curis) for suggestions. We thank M. Scott and Y.-C. Hu from the Gene Editing Core at CCHMC for their assistance.

Author information

Authors and Affiliations

Authors

Contributions

M.A.S., G.S.C. and A.P. performed most of the experiments with assistance from L.C.B. A.P., J.B., N.S., K.K., K.C. V.S., and A.K. performed the splicing and isoform analysis. M.W., S.K., and I.D.C.F. provided essential reagents. T.D.B., S.G.-M., D.V.A., K.P., U.S. and A.V. provided patient samples and insight. M.A.S., K.K., J.B., A.V. and D.T.S. conceived the project, designed experiments, interpreted results and wrote the manuscript.

Corresponding authors

Correspondence to Jacqueline Boultwood, Amit Verma or Daniel T. Starczynowski.

Ethics declarations

Competing interests

A.V. has received research funding from GlaxoSmithKline, Incyte, MedPacto, Novartis, Curis and Eli Lilly and Company, has received compensation as a scientific advisor to Novartis, Stelexis Therapeutics, Acceleron Pharma and Celgene, and has equity ownership in Stelexis Therapeutics.

Additional information

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

Integrated supplementary information

Supplementary Fig. 1 Molecularly and clinically distinct subtypes of AML based on global and IRAK4 RNA isoform expression.

(a) Overview of RNA isoform switching analysis in AML with the corresponding figures and panels shown in parentheses. (b) Schematic representation of expression-level regulation versus isoform-level regulation of RNA isoforms in hypothetical samples. (c) Overall survival analysis of the 3 major groupings of AML (Group 1, blue; Group 2, red; Group 3, green) characterized by distinct patterns of differential isoform usage identified in Fig. 1b. Likelihood ratio test, Multivariate P = 0.06. (d) Exon usage of innate immune genes undergoing isoform switching in AML (Group 2) was correlated to overall survival of AML. Shown is the z-score correlation of IRAK4 exon usage to overall survival of AML patients. In red, exon 4 significantly correlates with worse overall survival of AML patients. (e) Relative expression of IRAK4-L to IRAK4-S in normal tissue (blue) relative to cancer tissue (red) for the indicated cancers analyzed in TCGA. Breast cancer (n = 1093); normal tissue (n = 112). Colon cancer (n = 285); normal tissue (n = 41). Lung cancer (n = 515); normal tissue (n = 59). The P value is indicated for each pair of samples and was determined by Welch’s t-test. (f) Sequence analysis of the IRAK4 exon 4 cassette using primers flanking exon junctions 3-4, 4-5, and 3–5 isolated from THP1 cells.

Supplementary Fig. 2 IRAK4-L and IRAK4-S activation of NF-κB and MAPK pathways.

(a) Pathway analysis of enriched genes in AML patients preferentially expressing IRAK4-L (left) or IRAK4-S (right). Shown are the significant signalling networks and relevant genes associated with IRAK4-L or IRAK4-S expression in AML. Data is adapted from Fig. 2a. (b, c) HEK293 cells transfected with empty vector (pcDNA3.1), pcDNA3.1-IRAK4-L, or pcDNA3.1-IRAK4-S and then immunoblotted for NF-κB (p-IRAK1, p-p65) and MAPK (p-JNK, p-ERK, p-p38) signalling. (Three independent experiments). (d) HL60 cells, which express primarily IRAK4-S, were transduced with empty vector (pMSCV-pGK-GFP) or IRAK4-L and then immunoblotted for the indicated proteins. (Two independent experiments). (e) TF1 and HL60 cells were immunoblotted for IRAK4 with C-terminal antibody that detects IRAK4-L and IRAK4-S after immunoprecipitation of MyD88.

Supplementary Fig. 3 Generation and characterization of THP1 cells deficient for IRAK4 isoforms.

(a) Schematic showing the exons targeted for CRISPR-Cas9 mediated editing of the human IRAK4 gene locus in THP1 cells from 2 independent clones. (b) Immunoblotting for IRAK4 (N-terminal antibody) in matched parental (THP1) and IRAK4-edited (THP1 IRAK4 KO) THP1 cells. (c) Parental and IRAK4 KO THP1 cells expressing were examined for leukaemic progenitor function in methylcellulose using independent clones. Representative images are shown on the left. Scale bar, 700 microns. Data is summarized from 3 biological replicates. Error bars represent mean +/- SD. Two-sided t-tests were used for statistical analyses. (d) Total cell counts were performed for THP1 and THP1-IRAK4 KO (clone 14) cells in liquid culture for the indicated days. (e) Viable cell counts (Trypan blue exclusion) were performed for THP1 and THP1-IRAK4 KO (clone 14) cells in liquid culture for the indicated days. (f) THP1 cells transduced with shRNAs targeting IRAK4-L (shIRAK4-L), IRAK4-L and shIRAK4-S (shIRAK4-L/S), or empty control were monitored in liquid culture by GFP expression. (g) Flow cytometric analysis of THP1 and THP1-IRAK4 KO (clone 14) cells stained with CD14-PE or CD34-APC. Dark grey histogram represents no antibody control. Representative FACS histogram from 2 independent biological replicates. Data represent the mean ± s.e.m.

Supplementary Fig. 4 IRAK4-L is required for leukaemic cell function in vitro and correlates with worse overall survival in MDS.

(a) THP1 cells were transduced with mCherry-expressing shRNAs targeting both IRAK4-L and IRAK4-S (shIRAK4-L/S), or control shRNA, sorted for mCherry expression. Isolated cells (mCherry positive) were then transduced with cDNAs expressing IRAK4-L (pMSCV-pGK-GFP), IRAK4-S, or empty vector and sorted for mCherry/GFP expression. Double positive cells were then immunoblotted with the N-terminal IRAK4 antibody (left panel) and C-terminal IRAK4 antibody (right panel). (b-c) The indicated cells from panel (A) were examined for leukaemic progenitor function in methylcellulose. Representative images are shown in (B). Data in (C) is from 3 biological replicates. Scale bar, 700 microns. Error bars represent mean +/- SEM. Two-sided t-tests were used for statistical analyses. (d) Kaplan-meier survival analysis of MDS patients stratified based on expression of high or low IRAK4-L expression. Data is adapted from Gerstung et al. (Nature Communications, 2015). Data represent the mean ± s.e.m.

Supplementary Fig. 5 Inhibition of IRAK4-L reduces signalling and results in apoptosis in U2AF1-mutant AML cells.

(a–c) K562-U2AF1-S34F cells were treated with 10 μM IRAK1/4-Inh for 1 or 2 hours and immunoblotted for NF-κB (p-IRAK1, p-IKK) and MAPK (p-ERK, p-p38, p-JNK) activation (three biological replicates).

Supplementary Fig. 6 Genetic or pharmacologic inhibition of IRAK4 results in differentiation of U2AF1-mutant MDS, but does not affect normal haematopoiesis.

(a) Immunoblot analysis of THP1 cells using the N-terminal IRAK4 antibody following transfection with a control siRNA (siCTL) or siIRAK4 after 24 hours. (b) BM cells from MDS1 following transfection with a control siRNA (siCTL) or siIRAK4 were plated in methylcellulose (for 14 days) and analyzed by flow cytometry for myeloid differentiation. (c, d) BM cells from MDS3 (C) or normal CD34+ (D) cells were treated with 10 μM CA-4948 in methylcellulose (14 days) and then analyzed by flow cytometry for erythroid differentiation. (e) NSG mice received 12.5 mg/kg of CA-4948 (n = 9 mice) or vehicle (PBS; n = 7 mice) for 5 days per week for 12 weeks. Complete blood counts were determined at the end of the study. Two-sided t-tests were used for statistical analyses. Data represent the mean ± s.e.m.

Supplementary Fig. 7

Unprocessed images of all gels and blots.

Supplementary information

Supplementary Information

Supplementary Figures 1–7, Supplementary Table titles and legends

Reporting Summary

Supplementary Table 1

Genes that undergo RNA isoform switching in AML.

Supplementary Table 2

Genes undergoing mRNA isoform switching with at least one differential exon inclusion/exclusion event that correlates with AML survival.

Supplementary Table 3

Isoforms that correlate with worst prognosis in AML.

Supplementary Table 4

Description of antibodies.

Supplementary Table 5

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Smith, M.A., Choudhary, G.S., Pellagatti, A. et al. U2AF1 mutations induce oncogenic IRAK4 isoforms and activate innate immune pathways in myeloid malignancies. Nat Cell Biol 21, 640–650 (2019). https://doi.org/10.1038/s41556-019-0314-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0314-5

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer