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:

A non-canonical Notch complex regulates adherens junctions and vascular barrier function

Abstract

The vascular barrier that separates blood from tissues is actively regulated by the endothelium and is essential for transport, inflammation, and haemostasis1. Haemodynamic shear stress plays a critical role in maintaining endothelial barrier function2, but how this occurs remains unknown. Here we use an engineered organotypic model of perfused microvessels to show that activation of the transmembrane receptor NOTCH1 directly regulates vascular barrier function through a non-canonical, transcription-independent signalling mechanism that drives assembly of adherens junctions, and confirm these findings in mouse models. Shear stress triggers DLL4-dependent proteolytic activation of NOTCH1 to expose the transmembrane domain of NOTCH1. This domain mediates establishment of the endothelial barrier; expression of the transmembrane domain of NOTCH1 is sufficient to rescue defects in barrier function induced by knockout of NOTCH1. The transmembrane domain restores barrier function by catalysing the formation of a receptor complex in the plasma membrane consisting of vascular endothelial cadherin, the transmembrane protein tyrosine phosphatase LAR, and the RAC1 guanidine-exchange factor TRIO. This complex activates RAC1 to drive assembly of adherens junctions and establish barrier function. Canonical transcriptional signalling via Notch is highly conserved in metazoans and is required for many processes in vascular development, including arterial–venous differentiation3, angiogenesis4 and remodelling5. We establish the existence of a non-canonical cortical NOTCH1 signalling pathway that regulates vascular barrier function, and thus provide a mechanism by which a single receptor might link transcriptional programs with adhesive and cytoskeletal remodelling.

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: NOTCH1 regulates shear stress-induced vascular barrier function.
Figure 2: The NOTCH1 transmembrane domain mediates barrier function through interaction with VE-cadherin.
Figure 3: NOTCH1 assembles a mechanosensory junctional complex involving LAR, TRIO, and RAC.

Similar content being viewed by others

References

  1. Mehta, D. & Malik, A. B. Signaling mechanisms regulating endothelial permeability. Physiol. Rev. 86, 279–367 (2006)

    Article  CAS  Google Scholar 

  2. Chiu, J.-J. & Chien, S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol. Rev. 91, 327–387 (2011)

    Article  Google Scholar 

  3. Lawson, N. D. et al. Notch signaling is required for arterial–venous differentiation during embryonic vascular development. Development 19, 3675–3683 (2001)

    Google Scholar 

  4. Hellström, M. et al. Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis. Nature 445, 776–780 (2007)

    Article  ADS  Google Scholar 

  5. Krebs, L. T. et al. Notch signaling is essential for vascular morphogenesis in mice. Genes Dev. 14, 1343–1352 (2000)

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Li, Y.-S. J., Haga, J. H. & Chien, S. Molecular basis of the effects of shear stress on vascular endothelial cells. J. Biomech. 38, 1949–1971 (2005)

    Article  Google Scholar 

  7. Hahn, C. & Schwartz, M. A. Mechanotransduction in vascular physiology and atherogenesis. Nat. Rev. Mol. Cell Biol. 10, 53–62 (2009)

    Article  CAS  Google Scholar 

  8. Ayata, C. & Ropper, A. H. Ischaemic brain oedema. J. Clin. Neurosci. 9, 113–124 (2002)

    Article  Google Scholar 

  9. Dongaonkar, R. M., Stewart, R. H., Geissler, H. J. & Laine, G. A. Myocardial microvascular permeability, interstitial oedema, and compromised cardiac function. Cardiovasc. Res. 87, 331–339 (2010)

    Article  CAS  Google Scholar 

  10. Garcia-Cardeña, G., Comander, J., Anderson, K. R., Blackman, B. R. & Gimbrone, M. A. Jr. Biomechanical activation of vascular endothelium as a determinant of its functional phenotype. Proc. Natl Acad. Sci. USA 98, 4478–4485 (2001)

    Article  ADS  Google Scholar 

  11. Chen, X., Gays, D., Milia, C. & Santoro, M. M. Cilia control vascular mural cell recruitment in vertebrates. Cell Reports 18, 1033–1047 (2017)

    Article  CAS  Google Scholar 

  12. Artavanis-Tsakonas, S., Rand, M. D. & Lake, R. J. Notch signaling: cell fate control and signal integration in development. Science 284, 770–776 (1999)

    Article  ADS  CAS  Google Scholar 

  13. Bray, S. J. Notch signalling in context. Nat. Rev. Mol. Cell Biol. 17, 722–735 (2016)

    Article  CAS  Google Scholar 

  14. Weng, A. P. et al. Growth suppression of pre-T acute lymphoblastic leukemia cells by inhibition of Notch signaling. Mol. Cell. Biol. 23, 655–664 (2003)

    Article  CAS  Google Scholar 

  15. Rizzo, V., Morton, C., DePaola, N., Schnitzer, J. E. & Davies, P. F. Recruitment of endothelial caveolae into mechanotransduction pathways by flow conditioning in vitro. Am. J. Physiol. Heart. Circ. Physiol. 285, H1720–H1729 (2003)

    Article  CAS  Google Scholar 

  16. Gordon, W. R. et al. Mechanical allostery: evidence for a force requirement in the proteolytic activation of Notch. Dev. Cell 33, 729–736 (2015)

    Article  CAS  Google Scholar 

  17. Mammoto, T. et al. Angiopoietin-1 requires p190 RHOGAP to protect against vascular leakage in vivo. J. Biol. Chem. 282, 23910–23918 (2007)

    Article  CAS  Google Scholar 

  18. Mumm, J. S. et al. A ligand-induced extracellular cleavage regulates γ-secretase-like proteolytic activation of NOTCH1. Mol. Cell 5, 197–206 (2000)

    Article  CAS  Google Scholar 

  19. Huppert, S. S. et al. Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1. Nature 405, 966–970 (2000)

    Article  ADS  CAS  Google Scholar 

  20. Spindler, V., Schlegel, N. & Waschke, J. Role of GTPases in control of microvascular permeability. Cardiovasc. Res. 87, 243–253 (2010)

    Article  CAS  Google Scholar 

  21. Timmerman, I. et al. A local VE-cadherin and TRIO-based signaling complex stabilizes endothelial junctions through RAC1. J. Cell Sci. 128, 3041–3054 (2015)

    Article  CAS  Google Scholar 

  22. Debant, A. et al. The multidomain protein TRIO binds the LAR transmembrane tyrosine phosphatase, contains a protein kinase domain, and has separate RAC-specific and RHO-specific guanine nucleotide exchange factor domains. Proc. Natl Acad. Sci. USA 93, 5466–5471 (1996)

    Article  ADS  CAS  Google Scholar 

  23. Le Gall, M., De Mattei, C. & Giniger, E. Molecular separation of two signaling pathways for the receptor, Notch. Dev. Biol. 313, 556–567 (2008)

    Article  CAS  Google Scholar 

  24. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012)

    Article  CAS  Google Scholar 

  25. Coon, B. G. et al. Intramembrane binding of VE-cadherin to VEGFR2 and VEGFR3 assembles the endothelial mechanosensory complex. J. Cell Biol. 208, 975–986 (2015)

    Article  CAS  Google Scholar 

  26. Stittrich, A. B. et al. Mutations in NOTCH1 cause Adams-Oliver syndrome. Am. J. Hum. Genet. 95, 275–284 (2014)

    Article  CAS  Google Scholar 

  27. McDaniell, R. et al. NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway. Am. J. Hum. Genet. 79, 169–173 (2006)

    Article  CAS  Google Scholar 

  28. Joutel, A. et al. NOTCH3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature 383, 707–710 (1996)

    Article  ADS  CAS  Google Scholar 

  29. Smith, D. C. et al. A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors. Clin. Cancer Res. 20, 6295–6303 (2014)

    Article  ADS  CAS  Google Scholar 

  30. Bentley, K. et al. The role of differential VE-cadherin dynamics in cell rearrangement during angiogenesis. Nat. Cell Biol. 16, 309–321 (2014)

    Article  CAS  Google Scholar 

  31. Adamson, R. H., Lenz, J. F. & Curry, F. E. Quantitative laser scanning confocal microscopy on single capillaries: permeability measurement. Microcirculation 1, 251–265 (1994)

    Article  CAS  Google Scholar 

  32. Sturn, A., Quackenbush, J. & Trajanoski, Z. Genesis: cluster analysis of microarray data. Bioinformatics 18, 207–208 (2002)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported in part by grants from the National Institutes of Health (EB00262, EB08396, UH3EB017103 and HL115553) and the National Science Foundation Center for Engineering MechanoBiology (CMMI15-48571). W.J.P. acknowledges support from a Ruth L. Kirchstein National Research Service Award (F32 HL129733) and from the NIH through the Organ Design and Engineering Training program (T32 EB16652), and M.L.K. acknowledges support from the Hartwell Foundation and the NIH through the Translational Research in Regenerative Medicine Training program (T32 EB005583). We thank P. A. Murphy and R. Wang for discussions of preliminary data and M. Schwartz for materials and discussions.

Author information

Authors and Affiliations

Authors

Contributions

W.J.P., M.L.K., and C.S.C. conceived the study and designed experiments. W.J.P. and M.L.K. performed all experiments and data analysis with assistance from J.E. (qPCR) and J.Y. (Notch1-knockout mice). Y.W., H.V. and K.K.H. provided Notch1-knockout mice. W.J.P., M.L.K. and C.S.C. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Christopher S. Chen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks H. Gerhardt, I. Geudens, E. Tzima, A. Yap, and the other anonymous reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Figure 1 Known permeability-modulating agents regulate barrier function in hEMVs.

ad, Diffusive permeability measured in hEMVs in static or flow conditions treated with 500 nM S1P for 1 h (a), 10 μM SU415 (VEGFR2 inhibitor) overnight (b), 1 U ml−1 thrombin for 15 min (c), or 40 ng ml−1 VEGF for 1 h (d). e, Western blots of cleaved NOTCH1 ICD (using N1 V1754 antibody) from lysates of endothelial cells treated with vehicle or 40 ng ml−1 VEGF for 1 h. All plots show mean ± s.e.m., n ≥ 3 hEMVs, **P < 0.01. Exact P and n values are shown in Source Data; western blot is representative of two independent experiments.

Source data

Extended Data Figure 2 Flow activates expression of mechanotransduction-associated gene networks.

Relative gene expression measured by qPCR. –ΔΔCt of GOI under flow normalized to static control is plotted as a heat map (n = 3 flow or static qPCR analyses from distinct hEMV sets; each column is representative of an independent experiment; *P < 0.05, **P < 0.01). Exact P and n values are shown in Source Data.

Source data

Extended Data Figure 3 Non-transcriptional NOTCH1 signalling regulates vascular barrier function.

a, Expression of NOTCH1-target genes HES1 and HEY1 and the genes for the NOTCH1 ligand (DLL4) and VE-cadherin (CDH5) measured by qPCR in endothelial cells treated with DAPT or DMSO-load control on DLL4-coated and control tissue culture plastic substrates. b, Fluorescent micrograph of DLL4-coated device before cell seeding (green, Alexa Fluor 488–collagen I; red, DLL4 immunofluorescence). c, Fluorescent micrograph of endothelial cells in hEMVs coated with DLL4 before cell seeding. d, Micrographs of GFP-infection control cells under flow. e, Gene expression of HES1, HEY1, DLL4, NOTCH1, and CDH5 measured by qPCR in endothelial cells expressing dominant-negative MAML (dnMAML) or infection control (GFP). f, Western blot validation of NOTCH1 CRISPR lines: Scramble, NOTCH1KO, TMD-ICD KO, and ICD KO. g, Fluorescent micrographs of CRISPR–Cas9 scramble control cells cultured in flow conditions. h, Fluorescent micrographs of scramble control and NOTCH1KO hEMVs cultured in static conditions, immunostained for VE-cadherin and labelled with phalloidin. i, Quantification of junctional area measured from micrographs of cells immunostained for VE-cadherin as described in h. j, Gene expression measured by qPCR in NOTCH1KO and scramble control cells. k, Quantification of cell number in field of view at 10× magnification of scramble or NOTCH1KO hEMVs cultured in static and flow conditions. l, Micrographs of nuclei as visualized by DAPI in scramble or NOTCH1KO hEMVs. For all plots, mean ± s.e.m., n ≥ 3 hEMVs, **P < 0.01. Exact P and n values are shown in Source Data; images are representative of at least three independent experiments.

Source data

Extended Data Figure 4 NOTCH1 is activated in response to shear stress by endocytosis of DLL4.

a, ICD cleavage as measured by western blot with an antibody specific to cleaved ICD (N1 V1754) in DLL4KO and scramble control endothelial cells cultured under flow. b, Fluorescent micrographs of DLL4KO and scramble control hEMVs cultured under flow conditions, immunostained for VE-cadherin and labelled with phalloidin. c, Quantification of junctional area measured from micrographs of cells immunostained for VE-cadherin. d, Fluorescence micrograph of endothelial cells expressing DLL4–HA in static plus DMSO, flow plus DMSO, and flow plus Dynasore conditions, stained for haemagglutinin and DAPI. e, Quantification of internalized DLL4–HA in endothelial cells under static plus DMSO, flow plus DMSO, and flow plus Dynasore conditions. Cells with more than one Alexa Fluor 488-positive punctum counted as positive for internalized DLL4–HA. f, Immunofluorescence of an endothelial cell expressing DLL4–HA cultured under flow and stained for haemagglutinin and NOTCH1 ECD and stained with DAPI. g, Diffusive permeability of 70-kDa dextran in cells treated with Dynasore hydrate or DMSO-load control and exposed to flow overnight. All plots show mean ± s.e.m., n ≥ 3 hEMVs, **P < 0.01. Exact P and n values are given in Source Data; images are representative of at least two independent experiments.

Source data

Extended Data Figure 5 DAPT and NOTCH1 depletion modulate vascular permeability in vivo.

a, Diffusive permeability of mouse dermal vasculature as a function of vessel diameter after 1 h of intravenously injected DMSO or DAPT (n = 15 vessels across 3 mice per condition). b, High magnification whole-mount micrographs of Evans blue in the mouse dermal vasculature. Fluorescent images are representative of three independent experiments.

Source data

Extended Data Figure 6 NOTCH1 regulates junctional stability through association with VE-cadherin.

a, Time-lapse images of cells expressing VE-cadherin–mApple in the presence of DAPT or DMSO load control demonstrate that adherens junctions disassemble after 30 min of exposure to DAPT, leading to macroscopic intercellular gaps (red arrows). b, Fluorescent micrographs of NOTCH1KO cells expressing TMD–ICD–mApple or TMD–ICD(V1754G)–mApple immunostained for cleaved NOTCH1 (ICD V1754) and stained with DAPI. c, Fluorescent micrographs of TMD–mApple expressed in CHD5KO or scramble control endothelial cells and immunostained for VE-cadherin. d, Western blot for NOTCH1 ICD and VE-cadherin in NOTCH1KO and CHD5KO endothelial cells. e, Western blot validation of NOTCH1 rescue constructs: mApple, TMD–mApple, ICD–TMD–mApple, and ICD–TMD(V1754G)–mApple. f, Immunoprecipitation of VE-cadherin from hMVEC-D cells treated with DMSO or DAPT. Co-immunoprecipitation of mechanosensory complex components was assessed by immunoblotting for NOTCH1 ICD, TRIO, and LAR. g. Western blot of VE-cadherin immunoprecipitations from NOTCH1KO cells expressing NOTCH1-TMD truncation constructs (6, 8 and 12 amino acids from the N terminus) fused to mApple. h, Western blot of VE-cadherin immunoprecipitations from NOTCH1KO cells expressing NOTCH1 TMD constructs with single and double point mutations within the TMD and fused to mApple. Images are representative of at least three independent experiments.

Extended Data Figure 7 DLL4 and the NOTCH1 mechanosensory complex are critical for increased RAC1 activity in response to shear stress.

a, Active RAC1 was isolated from hMVEC-D cell lysates treated with DMSO and DAPT (20 μM) using a pull-down assay with GST–PBD. b, Quantification of band intensity from a demonstrates a decrease (~30%) in RAC1 activity with DAPT treatment. c, Active RAC1 was isolated from hMVEC-D cell lysates from static or shear flow conditions using GST–PBD pull-down. d, Active RAC1 was isolated using GST–PBD pull-down from DLL4KO cell lysates under flow conditions. e, Active RAC1 was isolated using GST–PBD pull-down from NOTCH1KO, PTPRFKO and TRIOKO cell lysates under flow conditions. Mean ± s.e.m., n = 3 independent lysates, **P < 0.01. Exact P and n values are shown in Source Data; all images are representative of at least three independent experiments.

Source data

Extended Data Figure 8 NOTCH1 regulates VE-cadherin-interacting proteins to form the NOTCH1 mechanosensory complex.

a, Immunoprecipitation of VE-cadherin from NOTCH1KO and scramble control cells. Co-immunoprecipitation of candidate NOTCH1-dependent VE-cadherin effectors was assessed by immunoblotting for VE-PTP, VEGFR2 and LAR (85 kDa P subunit). b, Immunoprecipitation of TRIO from scramble control, NOTCH1KO, and PTPRFKO cells. Immunoblotting for VE-cadherin was used to assess impaired TRIO–VE-cadherin co-immunoprecipitation on depletion of NOTCH1 or LAR. c, Western blots of VE-cadherin immunoprecipitates of lysates from lungs of Cdh5-cre+:Notch1fl/fl and control Cdh5-cre:Notch1fl/fl mice. d, Western blots of Trio immunoprecipitates from lysates of lungs from Cdh5-cre+:Notch1fl/fl and control Cdh5-cre:Notch1fl/fl mice. e, Western blot of proximal interacting proteins extracted with streptavidin from hMVEC-D cells expressing BIRA–HA (BioID) or VE-cadherin–BIRA–HA (VE-BioID) that were treated with DLL4, DAPT or DMSO. f, Western blot of proximal interacting proteins extracted with streptavidin in hMVEC-D cells expressing VE-cadherin–BIRA–HA (VE-BioID) that were treated with DLL4, DAPT or DMSO. Images are representative of at least two independent experiments.

Extended Data Figure 9 The NOTCH1 mechanosensory complex stabilizes cell–cell junctions through activation of RAC1.

Flow induces endocytosis of DLL4, triggering the activation and cleavage of NOTCH1 ICD and ECD, which allows the NOTCH1 TMD to link the adaptor protein LAR with VE-cadherin and recruit TRIO to adherens junctions. The resulting complex activates RAC1, elaborates cortical actin, and stabilizes cell–cell junctions to establish barrier function.

Extended Data Figure 10 Supplementary methods.

a, Schematic and summary of methods for quantifying vascular permeability in vivo with intravital microscopy. b, Intensity as a function of distance along lines connecting the nucleus centroids of neighbouring cells. Blue circles are local maxima used to count the number of stress fibres per unit length, and the shaded region is the area under the peak corresponding to cortical actin, and is normalized against the total area under the curve for quantification. The graph is representative of three independent experiments.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-2 and the uncropped western blot scans with size marker indications. (PDF 2940 kb)

Life Sciences Reporting Summary (PDF 88 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Polacheck, W., Kutys, M., Yang, J. et al. A non-canonical Notch complex regulates adherens junctions and vascular barrier function. Nature 552, 258–262 (2017). https://doi.org/10.1038/nature24998

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature24998

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: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research