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.

  • Original Article
  • Published:

Chronic myleproliferative neoplasms

Loss of HIF-1α accelerates murine FLT-3ITD-induced myeloproliferative neoplasia

Abstract

Hypoxia-induced signaling is important for normal and malignant hematopoiesis. The transcription factor hypoxia-inducible factor-1α (HIF-1α) has a crucial role in quiescence and self-renewal of hematopoietic stem cells (HSCs), as well as leukemia-initiating cells (LICs) of acute myeloid leukemia and chronic myeloid leukemia. We have investigated the effect of HIF-1α loss on the phenotype and biology of FLT-3ITD-induced myeloproliferative neoplasm (MPN). Using transgenic mouse models, we show that deletion of HIF-1α leads to an enhanced MPN phenotype reflected by an increased number of white blood cells, more severe splenomegaly and decreased survival. The proliferative effect of HIF-1α loss is cell intrinsic as shown by transplantation into recipient mice. HSC loss and organ-specific changes in the number and percentage of long-term HSCs were the most pronounced effects on a cellular level after HIF-1α deletion. Furthermore, we found a metabolic hyperactivation of malignant cells in the spleen upon loss of HIF-1α. Some of our findings are in contrary to what has been previously described for the role of HIF-1α in other myeloid hematologic malignancies and question the potential of HIF-1α as a therapeutic target.

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
Figure 2
Figure 3
Figure 4
Figure 5

Similar content being viewed by others

References

  1. Nilsson SK, Johnston HM, Coverdale JA . Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches. Blood 2001; 97: 2293–2299.

    Article  CAS  Google Scholar 

  2. Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E, Kubota Y et al. Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell 2010; 7: 391–402.

    Article  CAS  Google Scholar 

  3. Takubo K, Nagamatsu G, Kobayashi CI, Nakamura-Ishizu A, Kobayashi H, Ikeda E et al. Regulation of glycolysis by Pdk functions as a metabolic checkpoint for cell cycle quiescence in hematopoietic stem cells. Cell Stem Cell 2013; 12: 49–61.

    Article  CAS  Google Scholar 

  4. Rehn M, Olsson A, Reckzeh K, Diffner E, Carmeliet P, Landberg G et al. Hypoxic induction of vascular endothelial growth factor regulates murine hematopoietic stem cell function in the low-oxygenic niche. Blood 2011; 118: 1534–1543.

    Article  CAS  Google Scholar 

  5. Guitart AV, Subramani C, Armesilla-Diaz A, Smith G, Sepulveda C, Gezer D et al. Hif-2alpha is not essential for cell-autonomous hematopoietic stem cell maintenance. Blood 2013; 122: 1741–1745.

    Article  CAS  Google Scholar 

  6. Semenza GL . Hypoxia-inducible factors in physiology and medicine. Cell 2012; 148: 399–408.

    Article  CAS  Google Scholar 

  7. Pedersen M, Lofstedt T, Sun J, Holmquist-Mengelbier L, Pahlman S, Ronnstrand L . Stem cell factor induces HIF-1alpha at normoxia in hematopoietic cells. Biochem Biophys Res Commun 2008; 377: 98–103.

    Article  CAS  Google Scholar 

  8. Kirito K, Fox N, Komatsu N, Kaushansky K . Thrombopoietin enhances expression of vascular endothelial growth factor (VEGF) in primitive hematopoietic cells through induction of HIF-1alpha. Blood 2005; 105: 4258–4263.

    Article  CAS  Google Scholar 

  9. Simsek T, Kocabas F, Zheng J, Deberardinis RJ, Mahmoud AI, Olson EN et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 2010; 7: 380–390.

    Article  CAS  Google Scholar 

  10. Wang Y, Liu Y, Malek SN, Zheng P, Yang L . Targeting HIF1alpha eliminates cancer stem cells in hematological malignancies. Cell Stem Cell 2011; 8: 399–411.

    Article  CAS  Google Scholar 

  11. Rouault-Pierre K, Lopez-Onieva L, Foster K, Anjos-Afonso F, Lamrissi-Garcia I, Serrano-Sanchez M et al. HIF-2alpha protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress. Cell Stem Cell 2013; 13: 549–563.

    Article  CAS  Google Scholar 

  12. Hisa T, Spence SE, Rachel RA, Fujita M, Nakamura T, Ward JM et al. Hematopoietic, angiogenic and eye defects in Meis1 mutant animals. EMBO J 2004; 23: 450–459.

    Article  CAS  Google Scholar 

  13. Mayerhofer M, Valent P, Sperr WR, Griffin JD, Sillaber C . BCR/ABL induces expression of vascular endothelial growth factor and its transcriptional activator, hypoxia inducible factor-1alpha, through a pathway involving phosphoinositide 3-kinase and the mammalian target of rapamycin. Blood 2002; 100: 3767–3775.

    Article  CAS  Google Scholar 

  14. Zhang H, Li H, Xi HS, Li S . HIF1alpha is required for survival maintenance of chronic myeloid leukemia stem cells. Blood 2012; 119: 2595–2607.

    Article  CAS  Google Scholar 

  15. Chu SH, Heiser D, Li L, Kaplan I, Collector M, Huso D et al. FLT3-ITD knockin impairs hematopoietic stem cell quiescence/homeostasis, leading to myeloproliferative neoplasm. Cell Stem Cell 2012; 11: 346–358.

    Article  CAS  Google Scholar 

  16. Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res 2000; 60: 4010–4015.

    CAS  Google Scholar 

  17. Kuhn R, Schwenk F, Aguet M, Rajewsky K . Inducible gene targeting in mice. Science 1995; 269: 1427–1429.

    Article  CAS  Google Scholar 

  18. Lee BH, Tothova Z, Levine RL, Anderson K, Buza-Vidas N, Cullen DE et al. FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia. Cancer Cell 2007; 12: 367–380.

    Article  CAS  Google Scholar 

  19. Li L, Piloto O, Nguyen HB, Greenberg K, Takamiya K, Racke F et al. Knock-in of an internal tandem duplication mutation into murine FLT3 confers myeloproliferative disease in a mouse model. Blood 2008; 111: 3849–3858.

    Article  CAS  Google Scholar 

  20. Mead AJ, Kharazi S, Atkinson D, Macaulay I, Pecquet C, Loughran S et al. FLT3-ITDs instruct a myeloid differentiation and transformation bias in lymphomyeloid multipotent progenitors. Cell Rep 2013; 3: 1766–1776.

    Article  CAS  Google Scholar 

  21. Mupo A, Celani L, Dovey O, Cooper JL, Grove C, Rad R et al. A powerful molecular synergy between mutant nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice. Leukemia 2013; 27: 1917–1920.

    Article  CAS  Google Scholar 

  22. Denko NC . Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat Rev Cancer 2008; 8: 705–713.

    Article  CAS  Google Scholar 

  23. Yu WM, Liu X, Shen J, Jovanovic O, Pohl EE, Gerson SL et al. Metabolic regulation by the mitochondrial phosphatase PTPMT1 is required for hematopoietic stem cell differentiation. Cell Stem Cell 2013; 12: 62–74.

    Article  CAS  Google Scholar 

  24. Tothova Z, Gilliland DG . FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell 2007; 1: 140–152.

    Article  CAS  Google Scholar 

  25. Ito K, Suda T . Metabolic requirements for the maintenance of self-renewing stem cells. Nat Rev Mol Cell Biol 2014; 15: 243–256.

    Article  CAS  Google Scholar 

  26. Velasco-Hernandez T, Hyrenius-Wittsten A, Rehn M, Bryder D, Cammenga J . HIF-1alpha can act as a tumor suppressor gene in murine acute myeloid leukemia. Blood 2014; 124: 3597–3607.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr D Gary Gilliland (University of Pennsylvania, Philadelphia, PA, USA) for kindly providing Flt-3ITD knock-in mice and Dr David Bryder (Lund University, Lund, Sweden) for helpful discussions and advice on flow cytometry. This work was supported by project grants from Barncancerfonden, Cancerfonden and SSF through the Hemato-Linné and StemTherapy consortium (Sweden) (to JC) and a postdoctoral grant from Barncancerfonden (Sweden) (to TV-H).

Author Contributions

TV-H designed the research, performed experiments, analyzed data, created figures and wrote the manuscript; DT performed experiments; JC conceived and supervised the project, designed the research and wrote the manuscript. All authors read and approved the final manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J Cammenga.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Leukemia website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Velasco-Hernandez, T., Tornero, D. & Cammenga, J. Loss of HIF-1α accelerates murine FLT-3ITD-induced myeloproliferative neoplasia. Leukemia 29, 2366–2374 (2015). https://doi.org/10.1038/leu.2015.156

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/leu.2015.156

This article is cited by

Search

Quick links