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:

Dipeptidylpeptidase 4 negatively regulates colony-stimulating factor activity and stress hematopoiesis

Abstract

Enhancement of hematopoietic recovery after radiation, chemotherapy, or hematopoietic stem cell (HSC) transplantation is clinically relevant. Dipeptidylpeptidase (DPP4) cleaves a wide variety of substrates, including the chemokine stromal cell-derived factor-1 (SDF-1). In the course of experiments showing that inhibition of DPP4 enhances SDF-1–mediated progenitor cell survival, ex vivo cytokine expansion and replating frequency, we unexpectedly found that DPP4 has a more general role in regulating colony-stimulating factor (CSF) activity. DPP4 cleaved within the N-termini of the CSFs granulocyte-macrophage (GM)-CSF, G-CSF, interleukin-3 (IL-3) and erythropoietin and decreased their activity. Dpp4 knockout or DPP4 inhibition enhanced CSF activities both in vitro and in vivo. The reduced activity of DPP4-truncated versus full-length human GM-CSF was mechanistically linked to effects on receptor-binding affinity, induction of GM-CSF receptor oligomerization and signaling capacity. Hematopoiesis in mice after radiation or chemotherapy was enhanced in Dpp4−/− mice or mice receiving an orally active DPP4 inhibitor. DPP4 inhibition enhanced engraftment in mice without compromising HSC function, suggesting the potential clinical utility of this approach.

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: Deletion or inhibition of Dpp4 enhances SDF-1's effects on survival and ex vivo expansion of HPCs.
Figure 2: Inhibition of DPP4 enhances the in vitro activity of selected CSFs with DPP4 truncation sites.
Figure 3: Influence of soluble DPP4 on activities of recombinant mouse CSFs in vitro, and effects of full-length and truncated CSFs alone and in combination on hematopoiesis in vivo in mice.
Figure 4: Influence of DPP4 inhibition on colony formation, receptor binding and signaling in the TF-1 factor-dependent human cell line and in CD34+ cord blood cells.
Figure 5: Modeling of the GM-CSF GM-CSFR interaction, and effects of DPP4 deficiency or inhibition on hematopoietic recovery.
Figure 6: Influence of DPP4 inhibition in vivo on engraftment of HSCs and of CXCR4 deficiency on sitagliptin-enhanced hematopoietic recovery.

Similar content being viewed by others

References

  1. Broxmeyer, H.E. & Smith, F.O. Cord blood hematopoietic cell transplantation. in Thomas' Hematopoietic Cell Transplantation, 4th edn. (eds. Appelbaum, F.R., Forman, S.J., Negrin, R.S. & Blume, K.G.) 559–576 (Wiley-Blackwell, West Sussex, UK, 2009).

  2. Shaheen, M. & Broxmeyer, H.E. The humoral regulation of hematopoiesis. in Hematology: Basic Principles and Practice, 5th edn. (eds. Hoffman, R. et al.) 253–275 (Elsevier Churchill Livingston, Philadelphia, 2009).

  3. Rasmussen, H.B., Branner, S., Wiberg, F.C. & Wagtmann, N. Crystal structure of human dipeptidyl peptidase IV/CD26 in complex with a substrate analog. Nat. Struct. Biol. 10, 19–25 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Christopherson, K.W., Hangoc, G. & Broxmeyer, H.E. Cell surface peptidase CD26/dipeptidylpeptidase IV regulates CXCL12/stromal cell-derived factor-1 alpha-mediated chemotaxis of human cord blood CD34+ progenitor cells. J. Immunol. 169, 7000–7008 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Christopherson, K.W., Hangoc, G., Mantel, C.R. & Broxmeyer, H.E. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science 305, 1000–1003 (2004).

    Article  CAS  PubMed  Google Scholar 

  6. Tian, C., Bagley, J., Forman, D. & Iacomini, J. Inhibition of CD26 peptidase activity significantly improves engraftment of retrovirally transduced hematopoietic progenitors. Gene Ther. 13, 652–658 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Peranteau, W.H. et al. CD26 inhibition enhances allogeneic donor-cell homing and engraftment after in utero hematopoietic-cell transplantation. Blood 108, 4268–4274 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wyss, B.K. et al. Enhanced homing and engraftment of fresh but not ex vivo cultured murine marrow cells in submyeloablated hosts following CD26 inhibition by Diprotin A. Exp. Hematol. 37, 814–823 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Campbell, T.B., Hangoc, G., Liu, Y., Pollok, K. & Broxmeyer, H.E. Inhibition of CD26 in human cord blood CD34+ cells enhances their engraftment of nonobese diabetic/severe combined immunodeficiency mice. Stem Cells Dev. 16, 347–354 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Christopherson, K.W., Paganessi, L.A., Napier, S. & Porecha, N.K. CD26 inhibition on CD34+ or lineage human umbilical cord blood donor hematopoietic stem cells/hematopoietic progenitor cells improves long-term engraftment into NOD/SCID/β2null immunodeficient mice. Stem Cells Dev. 16, 355–360 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Kawai, T. et al. Diprotin A infusion into nonobese diabetic/severe combined immunodeficiency mice markedly enhances engraftment of human mobilized CD34+ peripheral blood cells. Stem Cells Dev. 16, 361–370 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Peled, A. et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science 283, 845–848 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Campbell, T.B. & Broxmeyer, H.E. CD26 inhibition and hematopoiesis: a novel approach to enhance transplantation. Front. Biosci. 13, 1795–1805 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Lataillade, J.J. et al. Stromal cell-derived factor 1 regulates primitive hematopoiesis by suppressing apoptosis and by promoting G0/G1 transition in CD34+ cells: evidence for an autocrine/paracrine mechanism. Blood 99, 1117–1129 (2002).

    Article  CAS  PubMed  Google Scholar 

  15. Broxmeyer, H.E. et al. Transgenic expression of stromal cell-derived factor-1/CXC chemokine ligand 12 enhances myeloid progenitor cell survival/antiapoptosis in vitro in response to growth factor withdrawal and enhances myelopoiesis in vivo. J. Immunol. 170, 421–429 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Broxmeyer, H.E. et al. Stromal cell-derived factor-1/CXCL12 directly enhances survival/antiapoptosis of myeloid progenitor cells through CXCR4 and Gαi proteins and enhances engraftment of competitive, repopulating stem cells. J. Leukoc. Biol. 73, 630–638 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, Y. et al. Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines. Blood 99, 4307–4317 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Piacibello, W. et al. Lentiviral gene transfer and ex vivo expansion of human primitive stem cells capable of primary, secondary, and tertiary multilineage repopulation in NOD/SCID mice. Blood 100, 4391–4400 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Broxmeyer, H.E. et al. Hematopoietic stem/progenitor cells, generation of induced pluripotent stem cells, and isolation of endothelial progenitors from 21- to 23.5-year cryopreserved cord blood. Blood 117, 4773–4777 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Carow, C.E. et al. Human multipotential progenitor cells (CFU-GEMM) have extensive replating capacity for secondary CFU-GEMM: An effect enhanced by cord blood plasma. Blood 81, 942–949 (1993).

    Article  CAS  PubMed  Google Scholar 

  21. Broxmeyer, H.E. et al. SDF-1/CXCL12 enhances in vitro replating capacity of murine and human multipotential and macrophage progenitor cells. Stem Cells Dev. 16, 589–596 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Broxmeyer, H.E. et al. The comparative effects in vivo of recombinant murine interleukin-3, natural murine colony stimulating factor-1 and recombinant murine granulocyte-macrophage colony stimulating factor on myelopoiesis in mice. J. Clin. Invest. 79, 721–730 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Broxmeyer, H.E. et al. Synergistic myelopoietic action in vivo of combinations of purified natural murine colony stimulating factor-1, recombinant murine interleukin-3, and recombinant murine granulocyte-macrophage colony stimulating factor administered to mice. Proc. Natl. Acad. Sci. USA 84, 3871–3875 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Broxmeyer, H.E. et al. Effects in vivo of purified recombinant human activin and erythropoietin, alone and in combination, in mice. Int. J. Hematol. 54, 447–454 (1991).

    CAS  PubMed  Google Scholar 

  25. Kitamura, T. et al. Establishment and characterization of a unique human cell line that proliferates dependently on GM-CSF, IL-3, or erythropoietin. J. Cell. Physiol. 140, 323–334 (1989).

    Article  CAS  PubMed  Google Scholar 

  26. Woodcock, J.M. et al. A single tyrosine residue in the membrane-proximal domain of the granulocyte-macrophage colony-stimulating factor, interleukin (IL)-3, and IL-5 receptor common β-chain is necessary and sufficient for high affinity binding and signaling by all three ligands. J. Biol. Chem. 271, 25999–26006 (1996).

    Article  CAS  PubMed  Google Scholar 

  27. Woodcock, J.M. et al. Receptors of the cytokine superfamily: mechanisms of activation and involvement in disease. Baillieres Clin. Haematol. 10, 507–524 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Woodcock, J.M. et al. The functional basis of granulocyte-macrophage colony stimulating factor, interleukin-3 and interleukin-5 receptor activation, basic and clinical implications. Int. J. Biochem. Cell Biol. 31, 1017–1025 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Haman, A. et al. Molecular determinants of the granulocyte-macrophage colony–stimulating factor receptor complex assembly. J. Biol. Chem. 274, 34155–34163 (1999).

    Article  CAS  PubMed  Google Scholar 

  30. Niu, L. et al. Kinetic resolution of two mechanisms for high-affinity granulocyte-macrophage colony–stimulating factor binding to its receptor. Blood 94, 3748–3753 (1999).

    Article  CAS  PubMed  Google Scholar 

  31. Lopez, A.F. et al. Molecular basis of cytokine receptor activation. IUBMB Life 62, 509–518 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Guthridge, M.A. et al. The phosphoserine-585–dependent pathway of the GM-CSF/IL-3/IL-5 receptors mediates hematopoietic cell survival through activation of NF-κB and induction of bcl-2. Blood 103, 820–827 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Hansen, G. et al. The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation. Cell 134, 496–507 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Hercus, T.R. et al. The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood 114, 1289–1298 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Broxmeyer, H.E. Inhibition in vivo of mouse granulopoiesis by cell-free activity derived from human polymorphonuclear neutrophils. Blood 51, 889–901 (1978).

    Article  CAS  PubMed  Google Scholar 

  36. Chitteti, B.R. et al. Phenotypic characterization of hematopoietic stem cells. in Cord Blood: Biology, Transplantation, Banking, and Regulation (ed. Broxmeyer, H.E.) 75–86 (AABB Press, Bethesda, Maryland, 2011).

  37. Waterstrat, A. et al. 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  PubMed  PubMed Central  Google Scholar 

  38. Christopherson, K.W. et al. G-CSF– and GM-CSF–induced upregulation of CD26 peptidase downregulates the functional chemotactic response of CD34+. Exp. Hematol. 34, 1060–1068 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Kidd, S. et al. In vivo expansion of the megakaryocyte progenitor cell population in adult CD26-deficient mice. Exp. Hematol. 39, 580–590 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Zaruba, M.M. et al. Synergy between CD26/DPP-IV inhibition and G-CSF improves cardiac function after acute myocardial infarction. Cell Stem Cell 4, 313–323 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Cho, S.Y. et al. The effect of CXCL12 processing on CD34+ cell migration in myeloproliferative neoplasms. Cancer Res. 70, 3402–3410 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Pang, R. et al. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell 6, 603–615 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Nie, Y., Han, Y.-C. & Zou, Y.-R. CXCR4 is required for the quiescence of primitive hematopoietic cells. J. Exp. Med. 205, 777–783 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Gotoh, A. et al. Steel factor induces serine phosphorylation of Stat3 in human growth factor-dependent myeloid cell lines. Blood 88, 138–145 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Adolfsson, J. et al. Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 121, 295–306 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Bryder, D., Rossi, D.J. & Weissman, I.L. Hematopoietic stem cells: the paradigmatic tissue-specific stem cell. Am. J. Pathol. 169, 338–346 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chitteti, B.R. et al. Impact of interactions of cellular components of the bone marrow microenvironment on hematopoietic stem and progenitor cell function. Blood 115, 3239–3248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Christensen, J.L. & Weissman, I.L. Flk-2 is a marker in hematopoietic stem cell differentiation: A simple method to isolate long-term stem cells. Proc. Natl. Acad. Sci. USA 98, 14541–14546 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kiel, M.J., Yilmaz, O.H., Iwashita, T., Terhorst, C. & Morrison, S.J. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121 (2005).

    Article  CAS  PubMed  Google Scholar 

  51. Kondo, M. et al. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu. Rev. Immunol. 21, 759–806 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Osawa, M., Hanada, K., Hamada, H. & Nakauchi, H. Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Sitnicka, E. et al. Key role of flt3 ligand in regulation of the common lymphoid progenitor but not in maintenance of the hematopoietic stem cell pool. Immunity 17, 463–472 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Mantel, C. et al. Cells enter a unique intermediate 4N stage, not 4N-G1, after aborted mitosis. Cell Cycle 7, 484–492 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Kajiyama, H. et al. Prolonged survival and decreased invasive activity attributable to dipeptidylpeptidase IV overexpression in ovarian carcinoma. Cancer Res. 62, 2753–2757 (2002).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

These studies were supported by US Health Service National Institutes of Health (NIH) grants R01 HL056416, R01 HL067284 and R01 HL112669 and a Center of Excellence in Hematology grant P01 DK090948 to H.E.B., and HL69669 and HL96305 to L.M.P. J.H. was supported as a doctoral candidate on NIH T32 DK07519 (to H.E.B.) and as a postdoc on NIH T32 HL07910 (to H.E.B.); H.A.O. was supported as a postdoc on NIH T32 DK07519; S.M.-G. was supported as a doctoral candidate on NIH R25 GM079657 (to H.E.B.); and T.B.C. was supported as a doctoral candidate on NIH T32 DK07519. S.L.R. was supported by NIH T32 CA111198 for parts of this study. We thank Y.-R. Zhou, The Feinstein Insitute for Medical Research, for the Cre+Cxcr4Fl/Fl mice.

Author information

Authors and Affiliations

Authors

Contributions

H.E.B. initiated the study, supervised experiments, contributed to concepts and experimental design, scored experiments, interpreted experimental data, did data analysis and wrote the manuscript. J.H. supervised experiments, contributed to concepts and experimental design, conducted and scored experiments, interpreted experimental data, did data analysis and helped in the writing of the manuscript. H.A.O. supervised experiments, contributed to concepts and experimental design, conducted and scored experiments, interpreted experimental data, did data analysis and helped in the writing of the manuscript. C.M. contributed to concepts and experimental design, conducted and scored experiments, interpreted experimental data, did data analysis and helped in the writing of the manuscript. B.R.C. conducted and scored experiments. S.C. conducted experiments and did data analysis. S.M.-G. conducted and scored experiments and did data analysis. G.H. contributed to concepts and experimental design, conducted and scored experiments and did data analysis. S.F. contributed to concepts and experimental design. S.L.R. and X.O. conducted and scored experiments. J.S. contributed to concepts and experimental design. L.M.P. contributed to concepts and experimental design, interpreted experimental data and helped in the writing of the manuscript. E.F.S. contributed to concepts and experimental design and interpreted experimental data. T.B.C. contributed to concepts and experimental design.

Corresponding author

Correspondence to Hal E Broxmeyer.

Ethics declarations

Competing interests

H.E.B. is a founder of the CordUse family cord blood bank and a member of the Medical Scientific Advisory Board of CordUse.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Table 1 (PDF 248 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Broxmeyer, H., Hoggatt, J., O'Leary, H. et al. Dipeptidylpeptidase 4 negatively regulates colony-stimulating factor activity and stress hematopoiesis. Nat Med 18, 1786–1796 (2012). https://doi.org/10.1038/nm.2991

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2991

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