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:

Pharmacological inhibition of EGFR signaling enhances G-CSF–induced hematopoietic stem cell mobilization

Abstract

Mobilization of hematopoietic stem and progenitor cells (HSPCs) from bone marrow into peripheral blood by the cytokine granulocyte colony–stimulating factor (G-CSF) has become the preferred source of HSPCs for stem cell transplants1,2,3,4,5,6,7,8,9. However, G-CSF fails to mobilize sufficient numbers of stem cells in up to 10% of donors, precluding autologous transplantation in those donors or substantially delaying transplant recovery time2. Consequently, new regimens are needed to increase the number of stem cells in peripheral blood upon mobilization. Using a forward genetic approach in mice, we mapped the gene encoding the epidermal growth factor receptor (Egfr) to a genetic region modifying G-CSF–mediated HSPC mobilization. Amounts of EGFR in HSPCs inversely correlated with the cells' ability to be mobilized by G-CSF, implying a negative role for EGFR signaling in mobilization. In combination with G-CSF treatment, genetic reduction of EGFR activity in HSPCs (in waved-2 mutant mice) or treatment with the EGFR inhibitor erlotinib increased mobilization. Increased mobilization due to suppression of EGFR activity correlated with reduced activity of cell division control protein-42 (Cdc42), and genetic Cdc42 deficiency in vivo also enhanced G-CSF–induced mobilization. Our findings reveal a previously unknown signaling pathway regulating stem cell mobilization and provide a new pharmacological approach for improving HSPC mobilization and thereby transplantation outcomes.

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: Regulation of G-CSF–mediated mobilization is linked to a 5-Mbp interval on mouse chromosome 11 containing the Egfr locus.
Figure 2: EGF reduces G-CSF–induced mobilization of HSPCs.
Figure 3: Genetic and pharmacological inhibition of EGFR activity enhances G-CSF–mediated mobilization.
Figure 4: Cdc42 regulates G-CSF–mediated mobilization in response to EGFR signaling.

Similar content being viewed by others

Accession codes

Accessions

ArrayExpress

References

  1. Katayama, Y. et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 124, 407–421 (2006).

    Article  CAS  Google Scholar 

  2. Broxmeyer, H.E. et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J. Exp. Med. 201, 1307–1318 (2005).

    Article  CAS  Google Scholar 

  3. Adams, G.B. et al. Therapeutic targeting of a stem cell niche. Nat. Biotechnol. 25, 238–243 (2007).

    Article  CAS  Google Scholar 

  4. Carlo-Stella, C. et al. Use of recombinant human growth hormone (rhGH) plus recombinant human granulocyte colony-stimulating factor (rhG-CSF) for the mobilization and collection of CD34+ cells in poor mobilizers. Blood 103, 3287–3295 (2004).

    Article  CAS  Google Scholar 

  5. Bonig, H., Wundes, A., Chang, K.H., Lucas, S. & Papayannopoulou, T. Increased numbers of circulating hematopoietic stem/progenitor cells are chronically maintained in patients treated with the CD49d blocking antibody natalizumab. Blood 111, 3439–3441 (2008).

    Article  CAS  Google Scholar 

  6. Cancelas, J.A. et al. Rac GTPases differentially integrate signals regulating hematopoietic stem cell localization. Nat. Med. 11, 886–891 (2005).

    Article  CAS  Google Scholar 

  7. Grigg, A.P. et al. Optimizing dose and scheduling of filgrastim (granulocyte colony–stimulating factor) for mobilization and collection of peripheral blood progenitor cells in normal volunteers. Blood 86, 4437–4445 (1995).

    CAS  PubMed  Google Scholar 

  8. Flomenberg, N. et al. The use of AMD3100 plus G-CSF for autologous hematopoietic progenitor cell mobilization is superior to G-CSF alone. Blood 106, 1867–1874 (2005).

    Article  CAS  Google Scholar 

  9. Breems, D.A. et al. Individual stem cell quality in leukapheresis products is related to the number of mobilized stem cells. Blood 87, 5370–5378 (1996).

    CAS  PubMed  Google Scholar 

  10. Geiger, H., Szilvassy, S.J., Ragland, P. & Van Zant, G. Genetic analysis of progenitor cell mobilization by granulocyte colony-stimulating factor: verification and mechanisms for loci on murine chromosomes 2 and 11. Exp. Hematol. 32, 60–67 (2004).

    Article  CAS  Google Scholar 

  11. Xing, Z. et al. Increased hematopoietic stem cell mobilization in aged mice. Blood 108, 2190–2197 (2006).

    Article  CAS  Google Scholar 

  12. Purton, L.E. & Scadden, D.T. Limiting factors in murine hematopoietic stem cell assays. Cell Stem Cell 1, 263–270 (2007).

    Article  CAS  Google Scholar 

  13. Waterstrat, A., Liang, Y., Swiderski, C.F., Shelton, B.J. & Van Zant, G. Congenic interval of CD45/Ly-5 congenic mice contains multiple genes that may influence hematopoietic stem cell engraftment. Blood 115, 408–417 (2010).

    Article  CAS  Google Scholar 

  14. Luetteke, N.C. et al. The mouse waved-2 phenotype results from a point mutation in the EGF receptor tyrosine kinase. Genes Dev. 8, 399–413 (1994).

    Article  CAS  Google Scholar 

  15. Liu, F., Poursine-Laurent, J. & Link, D.C. Expression of the G-CSF receptor on hematopoietic progenitor cells is not required for their mobilization by G-CSF. Blood 95, 3025–3031 (2000).

    CAS  PubMed  Google Scholar 

  16. Fabian, M.A. et al. A small molecule–kinase interaction map for clinical kinase inhibitors. Nat. Biotechnol. 23, 329–336 (2005).

    Article  CAS  Google Scholar 

  17. Malliri, A. et al. The transcription factor AP-1 is required for EGF-induced activation of rho-like GTPases, cytoskeletal rearrangements, motility and in vitro invasion of A431 cells. J. Cell Biol. 143, 1087–1099 (1998).

    Article  CAS  Google Scholar 

  18. Tu, S., Wu, W.J., Wang, J. & Cerione, R.A. Epidermal growth factor–dependent regulation of Cdc42 is mediated by the Src tyrosine kinase. J. Biol. Chem. 278, 49293–49300 (2003).

    Article  CAS  Google Scholar 

  19. Hall, A. Rho GTPases and the control of cell behaviour. Biochem. Soc. Trans. 33, 891–895 (2005).

    Article  CAS  Google Scholar 

  20. Yang, F.C. et al. Rac and Cdc42 GTPases control hematopoietic stem cell shape, adhesion, migration and mobilization. Proc. Natl. Acad. Sci. USA 98, 5614–5618 (2001).

    Article  CAS  Google Scholar 

  21. Yang, L. et al. Rho GTPase Cdc42 coordinates hematopoietic stem cell quiescence and niche interaction in the bone marrow. Proc. Natl. Acad. Sci. USA 104, 5091–5096 (2007).

    Article  CAS  Google Scholar 

  22. Natarajan, A., Wagner, B. & Sibilia, M. The EGF receptor is required for efficient liver regeneration. Proc. Natl. Acad. Sci. USA 104, 17081–17086 (2007).

    Article  CAS  Google Scholar 

  23. Seo, M. et al. Cdc42-dependent mediation of UV-induced p38 activation by G protein βγ subunits. J. Biol. Chem. 279, 17366–17375 (2004).

    Article  CAS  Google Scholar 

  24. Papayannopoulou, T. & Scadden, D.T. Stem-cell ecology and stem cells in motion. Blood 111, 3923–3930 (2008).

    Article  CAS  Google Scholar 

  25. Christopher, M.J., Liu, F., Hilton, M.J., Long, F. & Link, D.C. Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization. Blood 114, 1331–1339 (2009).

    Article  CAS  Google Scholar 

  26. Christopherson, K.W. II, Cooper, S. & Broxmeyer, H.E. Cell surface peptidase CD26/DPPIV mediates G-CSF mobilization of mouse progenitor cells. Blood 101, 4680–4686 (2003).

    Article  CAS  Google Scholar 

  27. Lévesque, J.P., Leavesley, D.I., Niutta, S., Vadas, M. & Simmons, P.J. Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins. J. Exp. Med. 181, 1805–1815 (1995).

    Article  Google Scholar 

  28. Yamaguchi, M. et al. Different adhesive characteristics and VLA-4 expression of CD34+ progenitors in G0/G1 versus S+G2/M phases of the cell cycle. Blood 92, 842–848 (1998).

    CAS  PubMed  Google Scholar 

  29. Glimm, H., Oh, I.H. & Eaves, C.J. Human hematopoietic stem cells stimulated to proliferate in vitro lose engraftment potential during their S/G(2)/M transit and do not reenter G(0). Blood 96, 4185–4193 (2000).

    CAS  PubMed  Google Scholar 

  30. Huygen, S. et al. Adhesion of synchronized human hematopoietic progenitor cells to fibronectin and vascular cell adhesion molecule-1 fluctuates reversibly during cell cycle transit in ex vivo culture. Blood 100, 2744–2752 (2002).

    Article  CAS  Google Scholar 

  31. Giet, O. et al. Increased binding and defective migration across fibronectin of cycling hematopoietic progenitor cells. Blood 99, 2023–2031 (2002).

    Article  CAS  Google Scholar 

  32. Lévesque, J.P., Takamatsu, Y., Nilsson, S.K., Haylock, D.N. & Simmons, P.J. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony–stimulating factor. Blood 98, 1289–1297 (2001).

    Article  Google Scholar 

  33. Semerad, C.L. et al. G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow. Blood 106, 3020–3027 (2005).

    Article  CAS  Google Scholar 

  34. Roberts, A.W. et al. Genetic influences determining progenitor cell mobilization and leukocytosis induced by granulocyte colony–stimulating factor. Blood 89, 2736–2744 (1997).

    CAS  PubMed  Google Scholar 

  35. Hasegawa, M., Baldwin, T.M., Metcalf, D. & Foote, S.J. Progenitor cell mobilization by granulocyte colony–stimulating factor controlled by loci on chromosomes 2 and 11. Blood 95, 1872–1874 (2000).

    CAS  PubMed  Google Scholar 

  36. Buerger, H. et al. Length and loss of heterozygosity of an intron 1 polymorphic sequence of egfr is related to cytogenetic alterations and epithelial growth factor receptor expression. Cancer Res. 60, 854–857 (2000).

    CAS  PubMed  Google Scholar 

  37. Wang, L., Yang, L., Filippi, M.D., Williams, D.A. & Zheng, Y. Genetic deletion of Cdc42GAP reveals a role of Cdc42 in erythropoiesis and hematopoietic stem/progenitor cell survival, adhesion and engraftment. Blood 107, 98–105 (2006).

    Article  CAS  Google Scholar 

  38. Wang, L., Yang, L., Debidda, M., Witte, D. & Zheng, Y. Cdc42 GTPase-activating protein deficiency promotes genomic instability and premature aging-like phenotypes. Proc. Natl. Acad. Sci. USA 104, 1248–1253 (2007).

    Article  CAS  Google Scholar 

  39. Yang, L., Wang, L. & Zheng, Y. Gene targeting of Cdc42 and Cdc42GAP affirms the critical involvement of Cdc42 in filopodia induction, directed migration, and proliferation in primary mouse embryonic fibroblasts. Mol. Biol. Cell 17, 4675–4685 (2006).

    Article  CAS  Google Scholar 

  40. Yuan, R., Astle, C.M., Chen, J. & Harrison, D.E. Genetic regulation of hematopoietic stem cell exhaustion during development and growth. Exp. Hematol. 33, 243–250 (2005).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported by a Cincinnati Children's Hospital Medical Center Translational Research Initiative Award to H.G. and M.A.R., grants from the US National Institutes of Health, HL076604 and DK077762 to H.G., AG024950 and HD060915 to G.V.Z. and an American Heart Association established investigator award to T.D.L.C. (0740069N). H.G. is a New Investigator in Aging, and G.V.Z. is a Senior Scholar in Aging of The Ellison Medical Foundation. A.S. is a recipient of the Lady Tata Memorial Trust Fellowship Award. We would like to thank T. Weaver and M.-D. Filippi for discussions and critical comments on the manuscript. We would like to thank B. Hardie for advice on the erlotinib experiments and the Comprehensive Mouse and Cancer Core as well as the Flow Cytometry Core at CCHMC for help with the experiments and OSI Pharmaceuticals for providing erlotinib.

Author information

Authors and Affiliations

Authors

Contributions

M.A.R. performed most of the experiments with help from K.J.N., A.S., D.S., D.D., W.L. and J.A.C. M.J. and A.W. performed microarray expression analyses. E.X. performed initial experiments on the inhibition of mobilization of EGF and generated the new congenic strains. J.A.C., N.R., T.D.L.C., G.V.Z., M.G., A.K. and Y.Z. consulted on most of the experiments and provided reagents or data for some experiments. H.G. performed some experiments and planned and supervised all experiments.

Corresponding author

Correspondence to Hartmut Geiger.

Ethics declarations

Competing interests

M.A.R. and H.G. are listed on a patent application [AU: About what aspect of this work?] to the US Patent Office filed by the Cincinnati Children's Hospital Medical Center.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7, Supplementary Tables 1 and 2 and Supplementary Methods (PDF 581 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ryan, M., Nattamai, K., Xing, E. et al. Pharmacological inhibition of EGFR signaling enhances G-CSF–induced hematopoietic stem cell mobilization. Nat Med 16, 1141–1146 (2010). https://doi.org/10.1038/nm.2217

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2217

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research