Skip to main content
Log in

Matrix deformations around angiogenic sprouts correlate to sprout dynamics and suggest pulling activity

  • Brief Communication
  • Published:
Angiogenesis Aims and scope Submit manuscript

Abstract

Angiogenesis is the formation of new blood vessels from the pre-existing vasculature. It is essential for normal tissue growth and regeneration, and also plays a key role in many diseases [Carmeliet in Nat Med 9:653–660, 2003]. Cytoskeletal components have been shown to be important for angiogenic sprout initiation and maintenance [Kniazeva and Putnam in Am J Physiol 297:C179–C187, 2009] as well as endothelial cell shape control during invasion [Elliott et al. in Nat Cell Biol 17:137–147, 2015]. The exact nature of cytoskeleton-mediated forces for sprout initiation and progression, however, remains poorly understood. Questions on the importance of tip cell pulling versus stalk cell pushing are to a large extent unanswered, which among others has to do with the difficulty of quantifying and resolving those forces in time and space. We developed methods based on time-lapse confocal microscopy and image processing—further termed 4D displacement microscopy—to acquire detailed, spatially and temporally resolved extracellular matrix (ECM) deformations, indicative of cell-ECM mechanical interactions around invading sprouts. We demonstrate that matrix deformations dependent on actin-mediated force generation are spatio-temporally correlated with sprout morphological dynamics. Furthermore, sprout tips were found to exert radially pulling forces on the extracellular matrix, which were quantified by means of a computational model of collagen ECM mechanics. Protrusions from extending sprouts mostly increase their pulling forces, while retracting protrusions mainly reduce their pulling forces. Displacement microscopy analysis further unveiled a characteristic dipole-like deformation pattern along the sprout direction that was consistent among seemingly very different sprout shapes—with oppositely oriented displacements at sprout tip versus sprout base and a transition zone of negligible displacements in between. These results demonstrate that sprout-ECM interactions are dominated by pulling forces and underline the key role of tip cell pulling for sprouting angiogenesis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Data availability

The data and code that support the findings of this study are available within the Supplementary Figures, Table, Videos, cited references or from the corresponding author upon request. The analytical code (together with a sample data set and guidelines) and laboratory protocols are available on the website of the corresponding author: https://www.mech.kuleuven.be/en/bme/research/mechbio, by navigating to the Software and Protocols section.

References

  1. Carmeliet P (2003) Angiogenesis in health and disease. Nat Med 9:653–660. https://doi.org/10.1038/nm0603-653

    Article  CAS  PubMed  Google Scholar 

  2. Kniazeva E, Putnam AJ (2009) Endothelial cell traction and ECM density influence both capillary morphogenesis and maintenance in 3-D. Am J Physiol 297:C179–C187. https://doi.org/10.1152/ajpcell.00018.2009

    Article  CAS  Google Scholar 

  3. Elliott H, Fischer RS, Myers KA et al (2015) Myosin II controls cellular branching morphogenesis and migration in three dimensions by minimizing cell-surface curvature. Nat Cell Biol 17:137–147. https://doi.org/10.1038/ncb3092

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gerhardt H (2008) VEGF and endothelial guidance in angiogenic sprouting. Organogenesis 4:241–246. https://doi.org/10.4161/org.4.4.7414

    Article  PubMed  PubMed Central  Google Scholar 

  5. Geudens I, Gerhardt H (2011) Coordinating cell behaviour during blood vessel formation. Development 138:4569–4583. https://doi.org/10.1242/dev.062323

    Article  CAS  PubMed  Google Scholar 

  6. Travasso RDM (2011) The mechanics of blood vessel growth. Vasculogenesis and angiogenesis - from embryonic development to regenerative medicine. InTechOpen, Rijeka, pp 187–204. https://doi.org/10.5772/34615

    Chapter  Google Scholar 

  7. Santos-Oliveira P, Correia A, Rodrigues T et al (2015) The force at the tip—modelling tension and proliferation in sprouting angiogenesis. PLOS Comput Biol 11:1–20. https://doi.org/10.1371/journal.pcbi.1004436

    Article  CAS  Google Scholar 

  8. De Smet F, Segura I, De Bock K et al (2009) Mechanisms of vessel branching: filopodia on endothelial tip cells lead the way. Arterioscler Thromb Vasc Biol 29:639–649. https://doi.org/10.1161/ATVBAHA.109.185165

    Article  CAS  PubMed  Google Scholar 

  9. Schmidt M, Paes K, De Mazière A et al (2007) EGFL7 regulates the collective migration of endothelial cells by restricting their spatial distribution. Development 134:2913–2923. https://doi.org/10.1242/dev.002576

    Article  CAS  PubMed  Google Scholar 

  10. Sauteur L, Krudewig A, Herwig L et al (2014) Cdh5/VE-cadherin promotes endothelial cell interface elongation via cortical actin polymerization during angiogenic sprouting. Cell Rep 9:504–513. https://doi.org/10.1016/j.celrep.2014.09.024

    Article  CAS  PubMed  Google Scholar 

  11. Kraning-Rush CM, Califano JP, Reinhart-King CA (2012) Cellular traction stresses increase with increasing metastatic potential. PLoS ONE 7:e32572. https://doi.org/10.1371/journal.pone.0032572

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Califano JP, Reinhart-King CA (2009) The effects of substrate elasticity on endothelial cell network formation and traction force generation. In: Soc MB (ed) Proceedings of the 31st Annual International Conference of the IEEE Engineering. Minneapolis, pp 3343–3345. https://doi.org/10.1109/IEMBS.2009.5333194

  13. Reinhart-King CA, Dembo M, Hammer DA (2005) The dynamics and mechanics of endothelial cell spreading. Biophys J 89:676–689. https://doi.org/10.1529/biophysj.104.054320

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Stout DA, Bar-Kochba E, Estrada JB et al (2016) Mean deformation metrics for quantifying 3D cell–matrix interactions without requiring information about matrix material properties. Proc Natl Acad Sci USA 113:2898–2903. https://doi.org/10.1073/pnas.1510935113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mabeta P, Pepper MS (2009) A comparative study on the anti-angiogenic effects of DNA-damaging and cytoskeletal-disrupting agents. Angiogenesis 12:81–90. https://doi.org/10.1007/s10456-009-9134-8

    Article  CAS  PubMed  Google Scholar 

  16. Yoon C, Choi C, Stapleton S et al (2019) Myosin IIA–mediated forces regulate multicellular integrity during vascular sprouting. Mol Biol Cell 30:1974–1984. https://doi.org/10.1091/mbc.e19-02-0076

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Du Y, Herath SCB, Wang Q-GG et al (2016) Three-dimensional characterization of mechanical interactions between endothelial cells and extracellular matrix during angiogenic sprouting. Sci Rep 6:1–14. https://doi.org/10.1038/srep21362

    Article  CAS  Google Scholar 

  18. Kniazeva E, Weidling JW, Singh R et al (2012) Quantification of local matrix deformations and mechanical properties during capillary morphogenesis in 3D. Integr Biol 4:431–439. https://doi.org/10.1039/c2ib00120a

    Article  CAS  Google Scholar 

  19. Du Y, Herath SCB, Wang Q-G et al (2012) Endothelial-cell-mediated displacement of extracellular matrix during angiogenesis. World Acad Sci Eng Technol 72:1541–1546. https://doi.org/10.5281/zenodo.1326688

    Article  Google Scholar 

  20. Steinwachs J, Metzner C, Skodzek K et al (2016) Three-dimensional force microscopy of cells in biopolymer networks. Nat Methods 13:171–176. https://doi.org/10.1038/nmeth.3685

    Article  CAS  PubMed  Google Scholar 

  21. Owen LM, Adhikari AS, Patel M et al (2017) A cytoskeletal clutch mediates cellular force transmission in a soft, three-dimensional extracellular matrix. Mol Biol Cell 28:1959–1974. https://doi.org/10.1091/mbc.E17-02-0102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Koch TM, Münster S, Bonakdar N et al (2012) 3D traction forces in cancer cell invasion. PLoS ONE 7:e33467. https://doi.org/10.1371/journal.pone.0033476

    Article  CAS  Google Scholar 

  23. Kim J, Jones CAR, Groves NS, Sun B (2016) Three-dimensional reflectance traction microscopy. PLoS ONE 11:1–17. https://doi.org/10.1371/journal.pone.0156797

    Article  CAS  Google Scholar 

  24. Jorge-Peñas A, Bové H, Sanen K et al (2017) 3D full-field quantification of cell-induced large deformations in fibrillar biomaterials by combining non-rigid image registration with label-free second harmonic generation. Biomaterials 136:86–97. https://doi.org/10.1016/j.biomaterials.2017.05.015

    Article  CAS  PubMed  Google Scholar 

  25. Condor M, Steinwachs J, Mark C et al (2017) Traction force microscopy in 3-dimensional extracellular matrix. Curr Protoc Cell Biol 75:1–20. https://doi.org/10.1002/cpcb.24

    Article  CAS  Google Scholar 

  26. Malandrino A, Trepat X, Kamm RD, Mak M (2019) Dynamic filopodial forces induce accumulation, damage, and plastic remodeling of 3D extracellular matrices. PLoS Comput Biol 15:1–26. https://doi.org/10.1371/journal.pcbi.1006684

    Article  CAS  Google Scholar 

  27. Kim J, Feng J, Jones CAR et al (2017) Stress-induced plasticity of dynamic collagen networks. Nat Commun 8:1–7. https://doi.org/10.1038/s41467-017-01011-7

    Article  CAS  Google Scholar 

  28. Kirkpatrick ND, Andreou S, Hoying JB, Utzinger U (2007) Live imaging of collagen remodeling during angiogenesis. Am J Physiol Heart Circ Physiol 292:H3198–H3206. https://doi.org/10.1152/ajpheart.01234.2006

    Article  CAS  PubMed  Google Scholar 

  29. Legant WR, Miller JS, Blakely BL et al (2010) Measurement of mechanical tractions exerted by cells within three-dimensional matrices. Nat Methods 7:969–971. https://doi.org/10.1038/nmeth.1531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bayless KJ, Kwak H-I, Su S-C (2009) Investigating endothelial invasion and sprouting behavior in three-dimensional collagen matrices. Nat Protoc 4:1888–1898. https://doi.org/10.1038/nprot.2009.221

    Article  CAS  PubMed  Google Scholar 

  31. Martins GG, Kolega J (2006) Endothelial cell protrusion and migration in three-dimensional collagen matrices. Cell Motil Cytoskeleton 63:101–115. https://doi.org/10.1002/cm.20104

    Article  PubMed  Google Scholar 

  32. Doyle AD, Carvajal N, Jin A et al (2015) Local 3D matrix microenvironment regulates cell migration through spatiotemporal dynamics of contractility-dependent adhesions. Nat Commun 6:8720. https://doi.org/10.1038/ncomms9720

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are grateful for funding support from the Research Foundation Flanders (FWO) (doctoral fellowship to T.H., postdoctoral fellowship to C.S., FWO grants G.0821.13, G0B9615N, G087018N), from the Hercules-foundation (G0H6316N), from KU Leuven internal funding (C14/17/111) and from the European Research Council under the European Union's Seventh Framework Program (FP7/2007–2013)/ ERC Grant Agreement No. 308223) to H.V.O. We are grateful to Anna Rita Cantelmo, Sandra Schoors, Inge Betz and Joris Souffreau for cell culture tips and techniques. We thank Evan Claes and Tobie Martens for their contributions to the experimental microscopy setup and the live imaging.

Author information

Authors and Affiliations

Authors

Contributions

HVO, MMV and AJP devised the overall experimental strategy. HVO and PC provided the cell culture facilities. MR provided the microscopy facilities. MMV extended the in vitro model for 4D displacement microscopy. CS operated and extended the microscopy setup for live cell imaging. MMV and CS performed the sprouting experiments, diffusion experiments and acellular control experiments. APJ. and JBF. developed all computational algorithms for displacement and morphology quantification. MMV and AJP pre-processed the data. JBF and MMV developed code for statistical analysing. MMV analysed the data, composed the main figures and rendered all videos. JBF developed computational algorithms for strain calculations and composed the figures related to strains. TH. developed the computational model of collagen mechanics and calculated sprout forces. MMV, AJP, JBF and HVO discussed and interpreted the data. The manuscript and figure design was prepared by MMV and was finalized with input from all authors.

Corresponding author

Correspondence to Hans Van Oosterwyck.

Ethics declarations

Conflict of interest

The authors declare no competing financial interests.

Additional information

Publisher's Note

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

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vaeyens, MM., Jorge-Peñas, A., Barrasa-Fano, J. et al. Matrix deformations around angiogenic sprouts correlate to sprout dynamics and suggest pulling activity. Angiogenesis 23, 315–324 (2020). https://doi.org/10.1007/s10456-020-09708-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10456-020-09708-y

Keywords

Navigation