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:

Targeting skeletal endothelium to ameliorate bone loss

This article has been updated

Abstract

Recent studies have identified a specialized subset of CD31hiendomucinhi (CD31hiEMCNhi) vascular endothelium that positively regulates bone formation. However, it remains unclear how CD31hiEMCNhi endothelium levels are coupled to anabolic bone formation. Mice with an osteoblast-specific deletion of Shn3, which have markedly elevated bone formation, demonstrated an increase in CD31hiEMCNhi endothelium. Transcriptomic analysis identified SLIT3 as an osteoblast-derived, SHN3-regulated proangiogenic factor. Genetic deletion of Slit3 reduced skeletal CD31hiEMCNhi endothelium, resulted in low bone mass because of impaired bone formation and partially reversed the high bone mass phenotype of Shn3−/− mice. This coupling between osteoblasts and CD31hiEMCNhi endothelium is essential for bone healing, as shown by defective fracture repair in SLIT3-mutant mice and enhanced fracture repair in SHN3-mutant mice. Finally, administration of recombinant SLIT3 both enhanced bone fracture healing and counteracted bone loss in a mouse model of postmenopausal osteoporosis. Thus, drugs that target the SLIT3 pathway may represent a new approach for vascular-targeted osteoanabolic therapy to treat bone loss.

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: Shn3−/− mice have higher levels of CD31hiEMCNhi endothelium.
Fig. 2: Ablation of Shn3 in osteoblasts enhances osteogenesis and angiogenesis in vivo.
Fig. 3: Inhibition of Shn3 enhances Slit3 expression in osteoblasts.
Fig. 4: Slit3−/− mice have reduced skeletal vasculature and bone mass in vivo.
Fig. 5: Osteoblast-derived Slit3 controls osteogenesis and CD31hiEMCNhi endothelium via ROBO1.
Fig. 6: Administration of recombinant SLIT3 has therapeutic effects on bone fracture healing and OVX-induced bone loss.

Similar content being viewed by others

Change history

  • 29 May 2018

    In the version of this article originally published, the Supplementary Information provided by the authors was not uploaded. The full version of the Supplementary Information now appears correctly.

References

  1. Kusumbe, A. P., Ramasamy, S. K. & Adams, R. H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 507, 323–328 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Ramasamy, S. K., Kusumbe, A. P., Wang, L. & Adams, R. H. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature 507, 376–380 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Xie, H. et al. PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat. Med. 20, 1270–1278 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Shim, J. H. et al. Schnurri-3 regulates ERK downstream of WNT signaling in osteoblasts. J. Clin. Invest. 123, 4010–4022 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Jones, D. C. et al. Regulation of adult bone mass by the zinc finger adapter protein Schnurri-3. Science 312, 1223–1227 (2006).

    Article  PubMed  CAS  Google Scholar 

  6. Nguyen Ba-Charvet, K. T. et al. Slit2-Mediated chemorepulsion and collapse of developing forebrain axons. Neuron 22, 463–473 (1999).

    Article  PubMed  CAS  Google Scholar 

  7. Jaworski, A. & Tessier-Lavigne, M. Autocrine/juxtaparacrine regulation of axon fasciculation by Slit-Robo signaling. Nat. Neurosci. 15, 367–369 (2012).

    Article  PubMed  CAS  Google Scholar 

  8. Jones, C. A. et al. Robo4 stabilizes the vascular network by inhibiting pathologic angiogenesis and endothelial hyperpermeability. Nat. Med. 14, 448–453 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Rama, N. et al. Slit2 signaling through Robo1 and Robo2 is required for retinal neovascularization. Nat. Med. 21, 483–491 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Vasam, G. et al. Reversal of bone marrow mobilopathy and enhanced vascular repair by angiotensin-(1-7) in diabetes. Diabetes 66, 505–518 (2017).

    Article  PubMed  CAS  Google Scholar 

  11. Geutskens, S. B. et al. Control of human hematopoietic stem/progenitor cell migration by the extracellular matrix protein Slit3. Lab. Invest. 92, 1129–1139 (2012).

    Article  PubMed  CAS  Google Scholar 

  12. Zhou, W. J., Geng, Z. H., Spence, J. R. & Geng, J. G. Induction of intestinal stem cells by R-spondin 1 and Slit2 augments chemoradioprotection. Nature 501, 107–111 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Mehlen, P., Delloye-Bourgeois, C. & Chedotal, A. Novel roles for Slits and netrins: axon guidance cues as anticancer targets? Nat. Rev. Cancer 11, 188–197 (2011).

    Article  PubMed  CAS  Google Scholar 

  14. Zhang, B. et al. Repulsive axon guidance molecule Slit3 is a novel angiogenic factor. Blood 114, 4300–4309 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Paul, J. D. et al. SLIT3-ROBO4 activation promotes vascular network formation in human engineered tissue and angiogenesis in vivo. J. Mol. Cell Cardiol. 64, 124–131 (2013).

    Article  PubMed  CAS  Google Scholar 

  16. Zhang, J. & Link, D. C. Targeting of mesenchymal stromal cells by Cre-recombinase transgenes commonly used to target osteoblast lineage cells. J. Bone Miner. Res. 31, 2001–2007 (2016).

    Article  PubMed  CAS  Google Scholar 

  17. Chen, J. et al. Osx-Cre targets multiple cell types besides osteoblast lineage in postnatal mice. PLoS ONE 9, e85161 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Su, A. I. et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proc. Natl Acad. Sci. USA 101, 6062–6067 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Yuan, W. et al. A genetic model for a central (septum transversum) congenital diaphragmatic hernia in mice lacking Slit3. Proc. Natl Acad. Sci. USA 100, 5217–5222 (2003).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Geutskens, S. B., Hordijk, P. L. & van Hennik, P. B. The chemorepellent Slit3 promotes monocyte migration. J. Immunol. 185, 7691–7698 (2010).

    Article  PubMed  CAS  Google Scholar 

  21. Naska, S., Lin, D. C., Miller, F. D. & Kaplan, D. R. p75NTR is an obligate signaling receptor required for cues that cause sympathetic neuron growth cone collapse. Mol. Cell Neurosci. 45, 108–120 (2010).

    Article  PubMed  CAS  Google Scholar 

  22. Howitt, J. A., Clout, N. J. & Hohenester, E. Binding site for Robo receptors revealed by dissection of the leucine-rich repeat region of Slit. EMBO J. 23, 4406–4412 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Liu, Z. et al. Extracellular Ig domains 1 and 2 of Robo are important for ligand (Slit) binding. Mol. Cell Neurosci. 26, 232–240 (2004).

    Article  PubMed  CAS  Google Scholar 

  24. Shin, M. et al. Vegfa signals through ERK to promote angiogenesis, but not artery differentiation. Development 143, 3796–3805 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Choi, H. J. et al. Yes-associated protein regulates endothelial cell contact-mediated expression of angiopoietin-2. Nat. Commun. 6, 6943 (2015).

    Article  PubMed  CAS  Google Scholar 

  26. Blockus, H. & Chedotal, A. Slit-Robo signaling. Development 143, 3037–3044 (2016).

    Article  PubMed  CAS  Google Scholar 

  27. Sakabe, M. et al. YAP/TAZ-CDC42 signaling regulates vascular tip cell migration. Proc. Natl Acad. Sci. USA 114, 10918–10923 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Bouxsein, M. L. et al. Ovariectomy-induced bone loss varies among inbred strains of mice. J. Bone Miner. Res. 20, 1085–1092 (2005).

    Article  PubMed  Google Scholar 

  29. Fukuda, T. et al. Sema3A regulates bone-mass accrual through sensory innervations. Nature 497, 490–493 (2013).

    Article  PubMed  CAS  Google Scholar 

  30. Zhang, Y. et al. Implant-derived magnesium induces local neuronal production of CGRP to improve bone-fracture healing in rats. Nat. Med. 22, 1160–1169 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Xu, R. Semaphorin 3A: a new player in bone remodeling. Cell Adh. Migr. 8, 5–10 (2014).

    Article  PubMed  Google Scholar 

  32. Ramalingam, P., Poulos, M. G. & Butler, J. M. Regulation of the hematopoietic stem cell lifecycle by the endothelial niche. Curr. Opin. Hematol. 24, 289–299 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  33. Buza, J. A. 3rd & Einhorn, T. Bone healing in 2016. Clin. Cases Miner. Bone Metab. 13, 101–105 (2016).

    PubMed  PubMed Central  Google Scholar 

  34. Sun, H., Dai, K., Tang, T. & Zhang, X. Regulation of osteoblast differentiation by slit2 in osteoblastic cells. Cells Tissues Organs 190, 69–80 (2009).

    Article  PubMed  Google Scholar 

  35. Leder, B. Z. et al. Denosumab and teriparatide transitions in postmenopausal osteoporosis (the DATA-Switch study): extension of a randomised controlled trial. Lancet 386, 1147–1155 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Wein, M. N. et al. Control of bone resorption in mice by Schnurri-3. Proc. Natl Acad. Sci. USA 109, 8173–8178 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chen, M. J., Yokomizo, T., Zeigler, B. M., Dzierzak, E. & Speck, N. A. Runx1 is required for the endothelial to haematopoietic cell transition but not thereafter. Nature 457, 887–891 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Rodda, S. J. & McMahon, A. P. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development 133, 3231–3244 (2006).

    Article  PubMed  CAS  Google Scholar 

  39. Lu, Y. et al. DMP1-targeted Cre expression in odontoblasts and osteocytes. J. Dent. Res. 86, 320–325 (2007).

    Article  PubMed  CAS  Google Scholar 

  40. Park, D. et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell 10, 259–272 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Xu, R. et al. c-Jun N-terminal kinases (JNKs) are critical mediators of osteoblast activity in vivo. J. Bone Miner. Res. 32, 1811–1815 (2017).

    Article  PubMed  CAS  Google Scholar 

  42. Tual-Chalot, S., Allinson, K.R., Fruttiger, M. & Arthur, H.M. Whole mount immunofluorescent staining of the neonatal mouse retina to investigate angiogenesis in vivo. J. Vis. Exp. 77, e50546 (2013).

    Google Scholar 

  43. Dempster, D. W. et al. Standardized nomenclature, symbols, and units for bone histomorphometry: a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. J. Bone Miner. Res. 28, 2–17 (2013).

    Article  PubMed  Google Scholar 

  44. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  PubMed  CAS  Google Scholar 

  45. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  PubMed  CAS  Google Scholar 

  46. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Zhang, C. et al. Mosaic: making biological sense of complex networks. Bioinformatics 28, 1943–1944 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Huang da, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  PubMed  CAS  Google Scholar 

  49. Greenblatt, M. B. et al. MEKK2 mediates an alternative beta-catenin pathway that promotes bone formation. Proc. Natl Acad. Sci. USA 113, E1226–E1235 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Bradaschia-Correa, V. et al. The selective serotonin reuptake inhibitor fluoxetine directly inhibits osteoblast differentiation and mineralization during fracture healing in mice. J. Bone Miner. Res. 32, 821–833 (2017).

    Article  PubMed  CAS  Google Scholar 

  51. Han, W. et al. The osteogenic potential of human bone callus. Sci. Rep. 6, 36330 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the many individuals who provided valuable reagents. M.B.G. holds a Career Award for Medical Scientists from the Burroughs Wellcome Foundation and is supported by the Office of the Director of the NIH under award DP5OD021351, a Junior Investigator Award from the Musculoskeletal Transplant Foundation and a March of Dimes Basil O’Connor Award. J.-H.S. is supported by National Institute of Arthritis and Musculoskeletal and Skin Diseases/NIH under R01AR068983 and a pilot project program award from UMass Center for Clinical and Translational Science. A.D.L. is supported by NIH/National Heart, Lung, and Blood Institute under R01 HL126913. This content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. We thank D. Ballon, B. He, B. -S. Ding and J. McCormick and Citigroup Biomedical Imaging Core, Weill Cornell Microscopy and Image Analysis and Flow Cytometry Core Facilities for technical support.

Author information

Authors and Affiliations

Authors

Contributions

R.X. and M.B.G. designed the experimental plan. R.X. executed most experiments. A.Y., Z.W., X.Y., P.K., N.L., Y.L., A.W., Y.Z., A.D.L., J.-H.S., J.M.B. and K.I. assisted with mouse studies. D.Y.S., J.-M.K., S.L. and B.Z. conducted in vitro experiments. S.D., G.J., H.D. and M.G.P. assisted with flow cytometry analysis and cell sorting. A.Q. collected human bone fracture samples. C.Z. performed RNA-seq analysis. M.P.B., B.Z. and J.-H.S. assisted with osteoclast studies. R.X. and M.B.G wrote the manuscript. M.B.G and L.H.G. supervised the project.

Corresponding authors

Correspondence to Laurie H. Glimcher or Matthew B. Greenblatt.

Ethics declarations

Competing interests

L.H.G. is on the board of directors of and holds equity in the GlaxoSmithKline and Waters Corporations. She is also a founder of Quentis Pharmaceuticals.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13 and Supplementary Table 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, R., Yallowitz, A., Qin, A. et al. Targeting skeletal endothelium to ameliorate bone loss. Nat Med 24, 823–833 (2018). https://doi.org/10.1038/s41591-018-0020-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0020-z

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