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:

Acute myeloid leukemia

Time resolved quantitative phospho-tyrosine analysis reveals Bruton’s Tyrosine kinase mediated signaling downstream of the mutated granulocyte-colony stimulating factor receptors

Abstract

Granulocyte-colony stimulating factor receptor (G-CSFR) controls myeloid progenitor proliferation and differentiation to neutrophils. Mutations in CSF3R (encoding G-CSFR) have been reported in patients with chronic neutrophilic leukemia (CNL) and acute myeloid leukemia (AML); however, despite years of research, the malignant downstream signaling of the mutated G-CSFRs is not well understood. Here, we used a quantitative phospho-tyrosine analysis to generate a comprehensive signaling map of G-CSF induced tyrosine phosphorylation in the normal versus mutated (proximal: T618I and truncated: Q741x) G-CSFRs. Unbiased clustering and kinase enrichment analysis identified rapid induction of phospho-proteins associated with endocytosis by the wild type G-CSFR only; while G-CSFR mutants showed abnormal kinetics of canonical Stat3, Stat5, and Mapk phosphorylation, and aberrant activation of Bruton’s Tyrosine Kinase (Btk). Mutant-G-CSFR-expressing cells displayed enhanced sensitivity (3–5-fold lower IC50) for ibrutinib-based chemical inhibition of Btk. Primary murine progenitor cells from G-CSFR-Q741x knock-in mice validated activation of Btk by the mutant receptor and retrovirally transduced human CD34+ umbilical cord blood cells expressing mutant receptors displayed enhanced sensitivity to Ibrutinib. A significantly lower clonogenic potential was displayed by both murine and human primary cells expressing mutated receptors upon ibrutinib treatment. Finally, a dramatic synergy was observed between ibrutinib and ruxolinitib at lower dose of the individual drug. Altogether, these data demonstrate the strength of unsupervised proteomics analyses in dissecting oncogenic pathways, and suggest repositioning Ibrutinib for therapy of myeloid leukemia bearing CSF3R mutations. Phospho-tyrosine data are available via ProteomeXchange with identifier PXD009662.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Touw IP, Palande K, Beekman R. Granulocyte colony-stimulating factor receptor signaling: implications for G-CSF responses and leukemic progression in severe congenital neutropenia. Hematol Oncol Clin North Am. 2013;27:61–73.

    Article  Google Scholar 

  2. Dwivedi P, Greis KD. Granulocyte colony-stimulating factor receptor signaling in severe congenital neutropenia, chronic neutrophilic leukemia, and related malignancies. Exp Hematol. 2017;46:9–20.

    Article  CAS  Google Scholar 

  3. Beekman R, Touw IP. G-CSF and its receptor in myeloid malignancy. Blood. 2010;115:5131–6.

    Article  CAS  Google Scholar 

  4. Rosenberg PS, Alter BP, Bolyard AA, Bonilla MA, Boxer LA, Chem B, et al. The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving long-term G-CSF therapy. Blood. 2006;107:4628–35.

    Article  CAS  Google Scholar 

  5. Gotlib J, Maxson JE, George TI, Tyner JW. The new genetics of chronic neutrophilic leukemia and atypical CML: implications for diagnosis and treatment. Blood. 2013;122:1707–11.

    Article  CAS  Google Scholar 

  6. Touw IP, Beekman R. Severe congenital neutropenia and chronic neutrophilic leukemia: an intriguing molecular connection unveiled by oncogenic mutations in CSF3R. Haematologica. 2013;98:1490–2.

    Article  CAS  Google Scholar 

  7. Zhang H, Nguyen-Jackson H, Panopoulos AD, Li HS, Murray PJ, Watowich SS. STAT3 controls myeloid progenitor growth during emergency granulopoiesis. Blood. 2010;116:2462–71.

    Article  CAS  Google Scholar 

  8. Beekman R, Valkhof MG, Sanders MA, van Strien PM, Haanstra JR, Broeders L, et al. Sequential gain of mutations in severe congenital neutropenia progressing to acute myeloid leukemia. Blood. 2012;119:5071–7.

    Article  CAS  Google Scholar 

  9. Mehta HM, Glaubach T, Long A, Lu H, Przychodzen B, Makishima H, et al. Granulocyte colony stimulating factor receptor T595I (T618I) mutation confers ligand independence and enhanced signaling. Leukemia. 2013;27:2407–10.

    Article  CAS  Google Scholar 

  10. Maxson JE, Gotlib J, Pollyea DA, Fleischman AG, Agarwal A, Eide CA, et al. Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. N Engl J Med. 2013;368:1781–90.

    Article  CAS  Google Scholar 

  11. Liu F, Kunter G, Krem MM, Eades WC, Cain JA, Tomasson MH, et al. Csf3r mutations in mice confer a strong clonal HSC advantage via activation of Stat5. J Clin Invest. 2008;118:946–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Pardanani A, Lasho TL, Laborde RR, Elliott M, Hanson CA, Knudson RA, et al. CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilic leukemia. Leukemia. 2013;27:1870–3.

    Article  CAS  Google Scholar 

  13. Dao KH, Solti MB, Maxson JE. Significant clinical response to JAK1/2 inhibition in a patient with CSF3R-T618I-positive atypical chronic myeloid leukemia. Leuk Res Rep. 2014;3:67–9.

    PubMed  PubMed Central  Google Scholar 

  14. Rohrabaugh S, Kesarwani M, Kincaid Z, Huber E, Leddonne J, Siddiqui Z, et al. Enhanced MAPK signaling is essential for CSF3R-induced leukemia. Leukemia. 2017;31:1770–8.

    Article  CAS  Google Scholar 

  15. Tony Hunter. Tyrosine phosphorylation: thirty years and counting. Curr Opin Cell Biol. 2009;21:140–6.

    Article  Google Scholar 

  16. Ong SE, Mann M. A practical recipe for stable isotope labeling by amino acids in cell culture (SILAC). Nat Protoc. 2006;1:2650–60.

    Article  CAS  Google Scholar 

  17. Rush J, Moritz A, Lee KA, Guo A, Goss VL, Spek EJ, et al. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells. Nat Biotechnol. 2005;23:94–101.

    Article  CAS  Google Scholar 

  18. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein–protein interaction networks, integrated over the tree of life. Nucleic Acid Res. 2015; 43:D447–52

  19. Lachmann A, Ma’ayan A. KEA: kinase enrichment analysis. Bioinformatics. 2009;25:684–6.

    Article  CAS  Google Scholar 

  20. Maxson JE, Luty SB, MacManiman JD, Abel ML, Druker BJ, Tyner JW. Ligand independence of the T618I mutation in the granulocyte colony-stimulating factor 3 receptor (CSF3R) protein results from loss of O-linked glycosylation and increased receptor dimerization. J Biol Chem. 2014;289:5820–7.

    Article  CAS  Google Scholar 

  21. Hermans MH, Antonissen C, Ward AC, Mayen AE, Ploemacher RE, Touw IP. Sustained receptor activation and hyperproliferation in response to granulocyte colony-stimulating factor (G-CSF) in mice with a severe congenital neutropenia/acute myeloid leukemiaderived mutation in the G-CSF receptor gene. J Exp Med. 1999;189:683–92.

    Article  CAS  Google Scholar 

  22. Aarts LH, Roovers O, Ward AC, Touw IP. Receptor activation and 2 distinct COOHterminal motifs control G-CSF receptor distribution and internalization kinetics. Blood. 2004;104:571–9.

    Article  Google Scholar 

  23. Wilson WH, Young RM, Schmitz RS, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–6.

    Article  CAS  Google Scholar 

  24. Rushworth SA, Murray MY, Zaitseva L, Bowles KM, MacEwan DJ. Identification of Bruton’s tyrosine kinase as a therapeutic target in acute myeloid leukemia. Blood. 2014;123:1229–38.

    Article  CAS  Google Scholar 

  25. Oellerich T, Mohr S, Corso J, Beck J, Dobele C, Braun H, et al. FLT3-ITD and TLR9 use Bruton Tyrosine kinase to activate distinct transcriptional programs mediating AML cell survival and proliferation. Blood. 2015;125:1936–47.

    Article  CAS  Google Scholar 

  26. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, et al. Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature. 2012;487:505–9.

    Article  CAS  Google Scholar 

  27. Stuible M, Abella JV, Feldhammer M, Nossov M, Sangwan V, Blagoev B, et al. Ptpb1b targets the endosomal sorting machinery: dephosphorylation of regulatory sites in the endosomal sorting complex required for transport component STAM2. J Biol Chem. 2010;285:23899–907.

    Article  CAS  Google Scholar 

  28. Wang L, Rudert WA, Loutaev I, Roginskaya V, Corey SJ. Repression of c-Cbl leads to enhanced G-CSF Jak-STAT signaling without increased cell proliferation. Oncogene. 2002;21:5346–55.

    Article  CAS  Google Scholar 

  29. Joffre C, Barrow R, Menard L, Calleja V, Hart IP, Kermogrant S. A direct role for Met endocytosis in tumorigenesis. Nat Cell Biol. 2011;13:827–37.

    Article  CAS  Google Scholar 

  30. Weinstein IB. Addiction to oncogenes-the Achilles heal of Cancer. Science. 2002;297:63–4.

    Article  CAS  Google Scholar 

  31. Kesharwani M, Kincaid Z, Gomaa A, Huber E, Rohrabaugh S, Siddiqui Z, et al. Trageting c-Fos and DUSP1 abrogates intrinsic resistance to tyrosine-kinase inhibitor therapy in BCR-ABL-induced leukemia. Nat Med. 2017;23:472–82.

    Article  Google Scholar 

  32. Khandanpour C, Phelan JD, Vassen L, Schutte J, Chen R, Horman SR, et al. Growth factor independence 1 antagonizes a p53-induced DNA damage response pathway in lymphoblastic leukemia. Cancer Cell. 2013;23:200–14.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr. Fan Dong (University of Toledo, Ohio) for providing 32D cell line and Dr. Dan Link (University of Washington, St. Louis, MO) for providing the truncation mutation G-CSFR mice. We are also grateful to Drs. Julia Maxson (Oregon Health Science University, Portland, OR) and D Ivo Touw (Erasmus Medical Center, Rotterdam, The Netherlands) for insightful suggestions during the study and Mr. Glenn Doermann for his expertize in graphic design for the figures. This article and our research work associated with G-CSFR are supported by several sources, including National Institutes of Health (NIH) Grant 1S10 RR027015-01 (KDG), the University of Cincinnati Millennium Scholars Fund (KDG), and the Cincinnati Children’s Hospital Research Foundation (KDG), Graduate Student Governance Association (GSGA) funding resources (PD), National Institutes of Health T32 ES007250-06 (DEM), as well as R01 CA196658 (HLG) and a grant from CancerFree Kids (HLG).

Author contributions

PD, DEM, MA, HLG, and KDG designed, performed, and analyzed all of the experiments. MA, HLG, and KDG provided funding, intellectual direction, and overall progression of the study. MW played an instrumental role in the data sorting and processing.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to H. Leighton Grimes or Kenneth D. Greis.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dwivedi, P., Muench, D.E., Wagner, M. et al. Time resolved quantitative phospho-tyrosine analysis reveals Bruton’s Tyrosine kinase mediated signaling downstream of the mutated granulocyte-colony stimulating factor receptors. Leukemia 33, 75–87 (2019). https://doi.org/10.1038/s41375-018-0188-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-018-0188-8

This article is cited by

Search

Quick links