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:

Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis

Abstract

Angiogenesis and the development of a vascular network are required for tumour progression, and they involve the release of angiogenic factors, including vascular endothelial growth factor (VEGF-A), from both malignant and stromal cell types1. Infiltration by cells of the myeloid lineage is a hallmark of many tumours, and in many cases the macrophages in these infiltrates express VEGF-A2. Here we show that the deletion of inflammatory-cell-derived VEGF-A attenuates the formation of a typical high-density vessel network, thus blocking the angiogenic switch in solid tumours in mice. Vasculature in tumours lacking myeloid-cell-derived VEGF-A was less tortuous, with increased pericyte coverage and decreased vessel length, indicating vascular normalization. In addition, loss of myeloid-derived VEGF-A decreases the phosphorylation of VEGF receptor 2 (VEGFR2) in tumours, even though overall VEGF-A levels in the tumours are unaffected. However, deletion of myeloid-cell VEGF-A resulted in an accelerated tumour progression in multiple subcutaneous isograft models and an autochthonous transgenic model of mammary tumorigenesis, with less overall tumour cell death and decreased tumour hypoxia. Furthermore, loss of myeloid-cell VEGF-A increased the susceptibility of tumours to chemotherapeutic cytotoxicity. This shows that myeloid-derived VEGF-A is essential for the tumorigenic alteration of vasculature and signalling to VEGFR2, and that these changes act to retard, not promote, tumour progression.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Deletion of VEGF in myeloid cells results in reduced vascularization but accelerated progression of mammary tumours.
Figure 2: Deletion of VEGF in myeloid cells leads to a normalized vasculature and higher tumour volumes in LLC.
Figure 3: Deletion of VEGF in myeloid cells results in reduced hypoxia and increased susceptibility of LLC tumours to cytotoxic agents.
Figure 4: Effect of tumour-cell-derived versus myeloid-cell-derived VEGF on tumour angiogenesis and growth.

Similar content being viewed by others

References

  1. Folkman, J. Angiogenesis. Annu. Rev. Med. 57, 1–18 (2006)

    Article  CAS  Google Scholar 

  2. Lewis, J. S., Landers, R. J., Underwood, J. C., Harris, A. L. & Lewis, C. E. Expression of vascular endothelial growth factor by macrophages is up-regulated in poorly vascularized areas of breast carcinomas. J. Pathol. 192, 150–158 (2000)

    Article  CAS  Google Scholar 

  3. Gerber, H. et al. VEGF couples hypertrophic cartilage remodeling, ossification and angiogenesis during endochondral bone formation. Nature Med. 5, 623–628 (1999)

    Article  CAS  Google Scholar 

  4. Clausen, B. E., Burkhardt, C., Reith, W., Renkawitz, R. & Forster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999)

    Article  CAS  Google Scholar 

  5. Cramer, T. et al. HIF-1α is essential for myeloid cell-mediated inflammation. Cell 112, 645–657 (2003)

    Article  CAS  Google Scholar 

  6. Guy, C. T., Cardiff, R. D. & Muller, W. J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol. Cell. Biol. 12, 954–961 (1992)

    Article  CAS  Google Scholar 

  7. Lin, E. Y. et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am. J. Pathol. 163, 2113–2126 (2003)

    Article  Google Scholar 

  8. Grunstein, J., Roberts, W. G., Mathieu-Costello, O., Hanahan, D. & Johnson, R. S. Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function. Cancer Res. 59, 1592–1598 (1999)

    CAS  PubMed  Google Scholar 

  9. Lin, E. Y. et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res. 66, 11238–11246 (2006)

    Article  CAS  Google Scholar 

  10. Mayer, E. L., Lin, N. U. & Burstein, H. J. Novel approaches to advanced breast cancer: bevacizumab and lapatinib. J. Natl. Compr. Canc. Netw. 5, 314–323 (2007)

    Article  CAS  Google Scholar 

  11. Hurwitz, H. I., Honeycutt, W., Haley, S. & Favaro, J. Long-term treatment with bevacizumab for patients with metastatic colorectal cancer: case report. Clin. Colorectal Cancer 6, 66–69 (2006)

    Article  Google Scholar 

  12. Gerber, H. P. et al. VEGF is required for growth and survival in neonatal mice. Development 126, 1149–1159 (1999)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. DuRose, N. Ferrara, L. Iruela-Arispe, L. Coussens, M. Karin, V. Nizet and H.-P. Gerber for helpful conversations. Financial support was provided by the Deutsche Forschungsgemeinschaft to C.S. (STO 787/1-1) and A.W. (WE 4275/1-1), a Susan G. Komen Dissertation Research Award (DISS0402406) to A.D., and National Institutes of Health grants CA82515, CA118165 and AI060840 to R.S.J.

Author Contributions C.S. generated isografts, conducted drug treatment studies, performed biochemical experiments, immunohistochemical procedures and histological analysis, and prepared the manuscript. A.D. generated MMTV-PyMT/LysMCre/VEGF+f/+f mice, conducted related tumour palpation/mass studies, initial histology and flow cytometry, and assisted with the manuscript. A.W. generated VEGF-null fibroblasts. N.Z. performed preliminary cDNA studies on MMTV-PyMT/LysMCre/VEGF+f/+f tumours. N.T. assisted with the fluorescein isothiocyanate-dextran angiography. J.I.G. and D.A.C. provided confocal microscopy images and technical advice. R.S.J. supervised and directed the project.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Randall S. Johnson.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-4 with Legends (PDF 6180 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Stockmann, C., Doedens, A., Weidemann, A. et al. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature 456, 814–818 (2008). https://doi.org/10.1038/nature07445

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature07445

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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