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:

Sulfated glycans engage the Ang–Tie pathway to regulate vascular development

Abstract

The angiopoietin (Ang)–Tie pathway is essential for the proper maturation and remodeling of the vasculature. Despite its importance in disease, the mechanisms that control signal transduction through this pathway are poorly understood. Here, we demonstrate that heparan sulfate glycosaminoglycans (HS GAGs) regulate Ang–Tie signaling through direct interactions with both Ang ligands and Tie1 receptors. HS GAGs formed ternary complexes with Ang1 or Ang4 and Tie2 receptors, resulting in potentiation of endothelial survival signaling. In addition, HS GAGs served as ligands for the orphan receptor Tie1. The HS–Tie1 interaction promoted Tie1–Tie2 heterodimerization and enhanced Tie1 stability within the mature vasculature. Loss of HS–Tie1 binding using CRISPR–Cas9-mediated mutagenesis in vivo led to decreased Tie protein levels, pathway suppression and aberrant retinal vascularization. Together, these results reveal that sulfated glycans use dual mechanisms to regulate Ang–Tie signaling and are important for the development and maintenance of the vasculature.

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: HS GAGs engage Ang1 and Ang4 ligands to form ternary complexes with Tie2.
Fig. 2: Ang-HS binding positively regulates prosurvival signaling.
Fig. 3: The orphan receptor Tie1 binds to the trisulfated motif of HS.
Fig. 4: HS engages Tie1 within its first Ig-like domain (Ig1).
Fig. 5: Loss of the HS–Tie1 interaction decreases Tie1–Tie2 heterodimerization.
Fig. 6: Ablation of the HS–Tie1 interaction in vivo causes aberrant retinal vascularization and loss of endothelial prosurvival signaling.

Similar content being viewed by others

Data availability

Data generated or analyzed during this study are included in the article and related Supplementary Information or are available from the corresponding author on reasonable request. Publicly available data used in this study include the Tie2 crystal structure (PDB 2GY5), the Tie1 protein sequence (UniProt P35590), the Dec. 2011 murine genome assembly (GRCm38/mm10) and the CHOPCHOP gRNA design tool (https://chopchop.cbu.uib.no/). Source data are provided with this paper.

References

  1. Potente, M., Gerhardt, H. & Carmeliet, P. Basic and therapeutic aspects of angiogenesis. Cell 146, 873–887 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Carmeliet, P. & Jain, R. K. Molecular mechanisms and clinical applications of angiogenesis. Nature 473, 298–307 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Adams, R. H. & Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol. 8, 464–478 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Simons, M., Gordon, E. & Claesson-Welsh, L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 17, 611–625 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Beenken, A. & Mohammadi, M. The FGF family: biology, pathophysiology and therapy. Nat. Rev. Drug Discov. 8, 235–253 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Andrae, J., Gallini, R. & Betsholtz, C. Role of platelet-derived growth factors in physiology and medicine. Genes Dev. 22, 1276–1312 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Capila, I. & Linhardt, R. J. Heparin–protein interactions. Angew. Chem. Int. Ed. Engl. 41, 391–412 (2002).

    Article  PubMed  Google Scholar 

  8. Xu, D. & Esko, J. D. Demystifying heparan sulfate–protein interactions. Annu. Rev. Biochem. 83, 129–157 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Poulain, F. E. & Yost, H. J. Heparan sulfate proteoglycans: a sugar code for vertebrate development? Development 142, 3456–3467 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bishop, J. R., Schuksz, M. & Esko, J. D. Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature 446, 1030–1037 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Olczyk, P., Mencner, Ł. & Komosinska-Vassev, K. Diverse roles of heparan sulfate and heparin in wound repair. Biomed. Res. Int. 2015, 549417 (2015).

  12. Forsberg, E. & Kjellen, L. Heparan sulfate: lessons from knockout mice. J. Clin. Invest. 108, 175–180 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fuster, M. M. & Wang, L. Endothelial heparan sulfate in angiogenesis. Prog. Mol. Biol. Transl. Sci. 93, 179–212 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jakobsson, L. et al. Heparan sulfate in trans potentiates VEGFR-mediated angiogenesis. Dev. Cell 10, 625–634 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Xu, D., Fuster, M. M., Lawrence, R. & Esko, J. D. Heparan sulfate regulates VEGF165- and VEGF121-mediated vascular hyperpermeability. J. Biol. Chem. 286, 737–745 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Schlessinger, J. et al. Crystal structure of a ternary FGF–FGFR–heparin complex reveals a dual role for heparin in FGFR binding and dimerization. Mol. Cell 6, 743–750 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Pellegrini, L., Burke, D. F., von Delft, F., Mulloy, B. & Blundell, T. L. Crystal structure of fibroblast growth factor receptor ectodomain bound to ligand and heparin. Nature 407, 1029–1034 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Abramsson, A. et al. Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development. Genes Dev. 21, 316–331 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. van Wijk, X. M. R. & van Kuppevelt, T. H. Heparan sulfate in angiogenesis: a target for therapy. Angiogenesis 17, 443–462 (2014).

    PubMed  Google Scholar 

  20. Augustin, H. G., Koh, G. Y., Thurston, G. & Alitalo, K. Control of vascular morphogenesis and homeostasis through the angiopoietin–Tie system. Nat. Rev. Mol. Cell Biol. 10, 165–177 (2009).

    Article  CAS  PubMed  Google Scholar 

  21. Saharinen, P., Eklund, L. & Alitalo, K. Therapeutic targeting of the angiopoietin–Tie pathway. Nat. Rev. Drug Discov. 16, 635–661 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Sato, T. N. et al. Distinct roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. Nature 376, 70–74 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Puri, M. C., Rossant, J., Alitalo, K., Bernstein, A. & Partanen, J. The receptor tyrosine kinase Tie is required for integrity and survival of vascular endothelial cells. EMBO J. 14, 5884–5891 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kim, M. et al. Opposing actions of angiopoietin-2 on Tie2 signaling and FOXO1 activation. J. Clin. Invest. 126, 3511–3525 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Partanen, J. et al. A novel endothelial cell surface receptor tyrosine kinase with extracellular epidermal growth factor homology domains. Mol. Cell. Biol. 12, 1698–1707 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Saharinen, P. et al. Multiple angiopoietin recombinant proteins activate the Tie1 receptor tyrosine kinase and promote its interaction with Tie2. J. Cell Biol. 169, 239–243 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Seegar, T. C. M. et al. Tie1–Tie2 interactions mediate functional differences between angiopoietin ligands. Mol. Cell 37, 643–655 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Leppänen, V.-M., Saharinen, P. & Alitalo, K. Structural basis of Tie2 activation and Tie2–Tie1 heterodimerization. Proc. Natl Acad. Sci. USA 114, 4376–4381 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  29. Savant, S. et al. The orphan receptor Tie1 controls angiogenesis and vascular remodeling by differentially regulating Tie2 in tip and stalk cells. Cell Rep. 12, 1761–1773 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Korhonen, E. A. et al. Tie1 controls angiopoietin function in vascular remodeling and inflammation. J. Clin. Invest. 126, 3495–3510 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. D’Amico, G. et al. Tie1 deletion inhibits tumor growth and improves angiopoietin antagonist therapy. J. Clin. Invest. 124, 824–834 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. La Porta, S. et al. Endothelial Tie1-mediated angiogenesis and vascular abnormalization promote tumor progression and metastasis. J. Clin. Invest. 128, 834–845 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Brown, J. M. et al. A sulfated carbohydrate epitope inhibits axon regeneration after injury. Proc. Natl Acad. Sci. USA. 109, 4768–4773 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rogers, C. J. et al. Elucidating glycosaminoglycan–protein–protein interactions using carbohydrate microarray and computational approaches. Proc. Natl Acad. Sci. USA. 108, 9747–9752 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Pulsipher, A., Griffin, M. E., Stone, S. E. & Hsieh-Wilson, L. C. Long-lived engineering of glycans to direct stem cell fate. Angew. Chem. Int. Ed. Engl. 54, 1466–1470 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Powell, A. K., Fernig, D. G. & Turnbull, J. E. Fibroblast growth factor receptors 1 and 2 interact differently with heparin/heparan sulfate: implications for dynamic assembly of a ternary signaling complex. J. Biol. Chem. 277, 28554–28563 (2002).

    Article  CAS  PubMed  Google Scholar 

  37. Teran, M. & Nugent, M. A. Synergistic binding of vascular endothelial growth factor-A and its receptors to heparin selectively modulates complex affinity. J. Biol. Chem. 290, 16451–16462 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Li, G. et al. Glycosaminoglycanomics of cultured cells using a rapid and sensitive LC-MS/MS approach. ACS Chem. Biol. 10, 1303–1310 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Griffith, A. R. et al. Predicting glycosaminoglycan surface protein interactions and implications for studying axonal growth. Proc. Natl Acad. Sci. USA. 114, 13697–13702 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Barton, W. A. et al. Crystal structures of the Tie2 receptor ectodomain and the angiopoietin-2–Tie2 complex. Nat. Struct. Mol. Biol. 13, 524–532 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Fredriksson, S. et al. Protein detection using proximity-dependent DNA ligation assays. Nat. Biotechnol. 20, 473–477 (2002).

    Article  CAS  PubMed  Google Scholar 

  42. Taichman, D. B. et al. A unique pattern of Tie1 expression in the developing murine lung. Exp. Lung Res. 29, 113–122 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Coles, C. H. et al. Proteoglycan-specific molecular switch for RPTPσ clustering and neuronal extension. Science 332, 484–488 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Meyer, R. D., Mohammadi, M. & Rahimi, N. A single amino acid substitution in the activation loop defines the decoy characteristic of VEGFR-1/FLT-1. J. Biol. Chem. 281, 867–875 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Vander Kooi, C. W. et al. Structural basis for ligand and heparin binding to neuropilin B domains. Proc. Natl Acad. Sci. USA 104, 6152–6157 (2007).

    Article  CAS  Google Scholar 

  46. Ibrahimi, O. A., Zhang, F., Hrstka, S. C. L., Mohammadi, M. & Linhardt, R. J. Kinetic model for FGF, FGFR and proteoglycan signal transduction complex assembly. Biochemistry 43, 4724–4730 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Griffin, M. E. & Hsieh-Wilson, L. C. Glycan engineering for cell and developmental biology. Cell Chem. Biol. 23, 108–121 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tully, S. E., Rawat, M. & Hsieh-Wilson, L. C. Discovery of a TNF-α antagonist using chondroitin sulfate microarrays. J. Am. Chem. Soc. 128, 7740–7741 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Waterhouse, A. et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296–W303 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mayo, S. L., Olafson, B. D. & Goddard, W. A. III DREIDING: a generic force field for molecular simulations. J. Phys. Chem. 94, 8897–8909 (1990).

    Article  CAS  Google Scholar 

  51. Kutner, R. H., Zhang, X.-Y. & Reiser, J. Production, concentration and titration of pseudotyped HIV-1-based lentiviral vectors. Nat. Protoc. 4, 495–505 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Montague, T. G., Cruz, J. M., Gagnon, J. A., Church, G. M. & Valen, E. CHOPCHOP: a CRISPR–Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 42, W401–W407 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mashiko, D. et al. Generation of mutant mice by pronuclear injection of circular plasmid expressing Cas9 and single guided RNA. Sci. Rep. 3, 3355 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR–Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Pease and staff of the Caltech Genetically Engineered Mouse Services Core for help with generating the Tie1-2A mouse line and J. Costanza and A. Gomez of the Caltech Office of Laboratory Animal Resources for mouse line care and maintenance. We also thank J. Vielmetter and the Caltech Protein Expression Center of the Beckman Institute for help with conducting the SPR experiments. This work was supported by the NIH (5R01GM093627 and 5R01GM127920 to L.C.H.-W.) and the National Science Foundation (CBET-1805022 to W.A.G., DGE-1144469 to M.E.G. and DGE-1745301 to A.W.S.).

Author information

Authors and Affiliations

Authors

Contributions

M.E.G. and L.C.H.-W. conceived the project. Unless otherwise noted, M.E.G. performed the experimental work. A.W.S. conducted some of the microarray, ELISA and cell imaging assays. G.M.M. aided in assay optimization for initial binding experiments and conducted all computational work under the guidance of W.A.G.III. All authors contributed to the design of the experimental and computational work and to data analysis, discussed the results and commented on the manuscript. M.E.G. and L.C.H.-W. wrote the manuscript. L.C.H.-W. supervised the project.

Corresponding author

Correspondence to Linda C. Hsieh-Wilson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data

Extended Data Fig. 1 Ang-Tie2 complex formation occurs in a sulfation-dependent manner.

Binding of biotinylated HS sulfation motifs to immobilized Tie2 in the presence or absence of a Ang1 or b Ang4, as detected by ELISA. Removal of the N- and O-sulfate groups (De-N,O), the N-sulfate groups (De-N), the O-sulfate groups (De-O), the 6-O-sulfate groups (De-6O), or the 2-O-sulfate groups (De-2O), or conversion of the N-sulfate to N-acetyl groups (N-Ac) reduced formation of HS-Ang1/4-Tie2 ternary complexes. Data represent mean ± s.e.m.; n = 2 independent replicates.

Source data

Extended Data Fig. 2 Orphan receptor Tie1 binds to the CS-E motif.

a, Chemical structure of the CS-E sulfation motif in CS polysaccharides. Interaction of CS-E and Tie1 by b ELISA, c glycan microarray, and d surface plasmon resonance. Dissociation constant for Tie1, as determined by ELISA: KD,app = 19.9 nM (10.5 to 53.5 nM); value represents mean (95% CI); graphed data represent n = 2 independent replicates. For glycan microarrays, data represent mean ± s.e.m.; n = 10 individual spots per glycan concentration. Dissociation constant for Tie1, as determined by SPR: KD,app = 14.7 nM (14.6 to 14.8 nM); value represents mean (95% CI). SPR data were fit using a 1:1 Langmuir model shown in red.

Source data

Extended Data Fig. 3 HS engages Tie1 in a sulfation-specific manner.

Binding of Tie1-Fc to different HS sulfation motifs, as determined by ELISA. Removal of the N- and O-sulfate groups (De-N,O), the O-sulfate groups (De-O), or the 6-O-sulfate groups (De-6O), or conversion of the N-sulfate to N-acetyl groups (N-Ac) abolished the HS–Tie1 interaction. Removal of the N-sulfate groups (De-N) or the 2-O-sulfate groups (De-2O) greatly reduced HS-Tie1 binding. Data represent mean ± s.e.m.; n = 2 independent replicates.

Source data

Extended Data Fig. 4 Soluble Tie1 binds to HUVECs.

Immunofluorescence imaging and orthogonal plane views of HUVECs with cell-associated Tie1-Fc (yellow), scale bar = 5 μm. Red bars indicate cross-sectional views in xz and yz images, which were produced by combining z-stack images using FIJI/ImageJ. The majority of Tie1 appears at the apical or basolateral cell surface. Representative images from four individual cells are shown.

Extended Data Fig. 5 Sulfated HS GAGs engage Tie1 in its N-terminal domains.

a Schematic and b Western blot of overexpressed, purified full-length (Tie1-FL) and C-terminal truncated (Tie-N) Tie1 ectodomains. Colors in (a) correspond to the ribbon diagram of Tie1 in Fig. 4a. Gray sections are derived from the expression plasmid. c, Binding of biotinylated triS-HS to recombinant Tie1-FL and Tie-N by ELISA. Data for (c) were obtained in the same experiment as Fig. 4e. Dissociation constants, as determined by ELISA: KD,app (Tie1-FL) = 6.51 nM (5.58 to 7.63 nM), KD,app (Tie1-N) = 6.26 nM (5.26 to 7.45 nM); values represent mean (95% CI); graphed data represent n = 2 independent replicates.

Source data

Extended Data Fig. 6 HS GAGs engage Tie1 in an electropositive region of the first N-terminal Ig-like domain.

a Ribbon diagram and b electrostatic potential surface of the Tie2 crystal structure (pdb 2GY5), which lacks the electropositive HS binding site found in Tie1. The red to blue scale in (b) represents relative electrostatic potential (electronegative to electropositive). c, Schematic of the first N-terminal Ig-like domain (Ig1) of Tie1, highlighting the six positively charged amino acids within 5-10 Å of the top 10 highest ranked HS GAG docked poses. All residues are mutated to alanine in the Tie1-6A construct, whereas only the two bolded residues are mutated in Tie1-2A. d, Structural model of the Tie1 N-terminal region (residues 22-447) indicating key amino acid residues involved in HS binding (purple) and those reported to be involved in Tie2 binding (orange)27. Distances are displayed in angstroms between the alpha carbon of each residue. e, Western blot showing expression of secreted Tie1 ectodomain constructs from HEK-293T cells after purification.

Source data

Extended Data Fig. 7 The HS-Tie1 interaction is driven primarily by ionic interactions.

a, Relative energetic contributions of van der Waals, hydrogen bonding, and Coulombic forces to the total calculated nonbonding energy are shown for the top 10 HS-Tie1 binding poses. In all cases, Coulombic forces dominated the calculated energy. b, Salt elution profile of Tie1 bound to heparin-sepharose shows loss of the HS-Tie1 interaction with increasing NaCl concentrations, indicative of strong ionic interactions between HS and Tie1. FT = flow-through.

Source data

Extended Data Fig. 8 Generation, genotyping, and characterization of Tie1-2A mice.

a, Schematic of the homology-directed repair method used to generate the Tie1-2A mouse line incorporating R38A and R82 A mutations into the endogenous murine Tie1 gene. b, Representative genotyping results from a Tie12A/wt x Tie12A/wt breeding pair after the Tie1 locus was amplified by PCR and digested with StuI. Animal 7 is Tie1wt/wt, animals 1, 2, 3, and 5 are Tie12A/wt, and animals 4 and 6 are Tie12A/2A. c, Quantification of weight differences between Tie12A/2A (2A/2A) and wild-type (wt/wt) 4-month-old, male littermates, n = 5 per genotype. d,e, Quantification of relative vessel area and branchpoints from retinal samples described in Fig. 6; n = 5 per genotype. Data represent mean ± s.e.m.; unpaired, two-tailed Student’s t test.

Source data

Extended Data Fig. 9 Tie1 mutation does not affect Erk1/2 phosphorylation in vivo.

a, Representative Western blots and b, quantification of pErk1/2 from lung tissue harvested from 7-day-old pups; n = 6 per genotype. pErk1/2 values were normalized to the corresponding total Erk1/2 levels and are reported relative to the wild-type animals. All gels were transferred to the same blot and imaged to allow for direct comparison between samples. Data represent mean ± s.e.m.; unpaired, two-tailed Student’s t test.

Source data

Extended Data Fig. 10 HS GAGs regulate the Ang/Tie signaling pathway.

HS GAGs utilize two distinct binding modes to promote pathway activation. a, Endothelial cell-associated HS GAGs recruit agonistic ligands Ang1 and Ang4 to form HS-Ang-Tie2 complexes and elicit downstream signaling. b, HS GAGs also bind to the orphan receptor Tie1 to promote the Tie1-Tie2 interaction, which maintains Tie2 protein levels and prevents protein turnover at the cell surface to sustain downstream signaling.

Supplementary information

Supplementary Information

Supplementary Figs. 1–4

Reporting Summary

Supplementary Data

Source Data for Supplementary Fig. 3

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data and unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data and unprocessed western blots.

Source Data Fig. 6

Statistical source data and unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data and unprocessed western blot.

Source Data Extended Data Fig. 6

Unprocessed western blot.

Source Data Extended Data Fig. 7

Statistical source data and unprocessed western blot.

Source Data Extended Data Fig. 8

Statistical source data and unprocessed DNA gel.

Source Data Extended Data Fig. 9

Statistical source data and unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Griffin, M.E., Sorum, A.W., Miller, G.M. et al. Sulfated glycans engage the Ang–Tie pathway to regulate vascular development. Nat Chem Biol 17, 178–186 (2021). https://doi.org/10.1038/s41589-020-00657-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-00657-7

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