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.

  • Protocol
  • Published:

Microfabrication of AngioChip, a biodegradable polymer scaffold with microfluidic vasculature

Abstract

Microengineered biomimetic systems for organ-on-a-chip or tissue engineering purposes often fail as a result of an inability to recapitulate the in vivo environment, specifically the presence of a well-defined vascular system. To address this limitation, we developed an alternative method to cultivate three-dimensional (3D) tissues by incorporating a microfabricated scaffold, termed AngioChip, with a built-in perfusable vascular network. Here, we provide a detailed protocol for fabricating the AngioChip scaffold, populating it with endothelial cells and parenchymal tissues, and applying it in organ-on-a-chip drug testing in vitro and surgical vascular anastomosis in vivo. The fabrication of the AngioChip scaffold is achieved by a 3D stamping technique, in which an intricate microchannel network can be embedded within a 3D scaffold. To develop a vascularized tissue, endothelial cells are cultured in the lumen of the AngioChip network, and parenchymal cells are encapsulated in hydrogels that are amenable to remodeling around the vascular network to form functional tissues. Together, these steps yield a functional, vascularized network in vitro over a 14-d period. Finally, we demonstrate the functionality of AngioChip-vascularized hepatic and cardiac tissues, and describe direct surgical anastomosis of the AngioChip vascular network on the hind limb of a Lewis rat model.

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
Fig. 2: POMaC polymer synthesis and purification.
Fig. 3: Microfabrication of AngioChip scaffold with 3D stamping technique.
Fig. 4: Assembly of an AngioChip bioreactor with seeding of endothelial cells and parenchymal tissues.
Fig. 5: Endothelialized lumen of AngioChip scaffold and sprouting of endothelial cells.
Fig. 6: Characterization of self-assembled tissues after 7 d of cultivation.
Fig. 7: Direct surgical anastomosis of vascularized cardiac AngioChip scaffolds to hind limbs of an adult Lewis rat.

Similar content being viewed by others

References

  1. Betts, J. G. et al. Anatomy and Physiology (Open Stax College, Houston, TX, 2013).

  2. Langer, R. & Vacanti, J. P. Tissue engineering. Science 260, 920–926 (1993).

    Article  PubMed  CAS  Google Scholar 

  3. Lovett, M., Lee, K., Edwards, A. & Kaplan, D. L. Vascularization strategies for tissue engineering. Tissue Eng. Part B Rev. 15, 353–370 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Atala, A., Kasper, F. K. & Mikos, A. G. Engineering complex tissues. Sci. Transl. Med. 4, 160rv112 (2012).

    Article  CAS  Google Scholar 

  5. Miller, J. S. The billion cell construct: will three-dimensional printing get us there? PLoS Biol. 12, e1001882 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Montgomery, M., Zhang, B. & Radisic, M. Cardiac tissue vascularization from angiogenesis to microfluidic blood vessels. J. Cardiovasc. Pharmacol. Ther. 19, 382–393 (2014).

    Article  PubMed  CAS  Google Scholar 

  7. Ingber, D. E. Reverse engineering human pathophysiology with organs-on-chips. Cell 164, 1105–1109 (2016).

    Article  PubMed  CAS  Google Scholar 

  8. Zhang, B. & Radisic, M. Organ-on-a-chip devices advance to market. Lab Chip 17, 2395–2420 (2017).

    Article  PubMed  CAS  Google Scholar 

  9. Esch, E. W., Bahinski, A. & Huh, D. Organs-on-chips at the frontiers of drug discovery. Nat. Rev. Drug Discov. 14, 248–260 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Takebe, T., Zhang, B. & Radisic, M. Synergistic engineering: organoids meet organs-on-a-chip. Cell Stem Cell 21, 297–300 (2017).

    Article  PubMed  CAS  Google Scholar 

  11. Ahadian, S. et al. Organ-on-a-chip platforms: a convergence of advanced materials, cells, and microscale technologies. Adv. Healthc. Mater. 7, 1700506 (2018).

  12. Young, E. W. K. & Simmons, C. A. Macro- and microscale fluid flow systems for endothelial cell biology. Lab Chip 10, 143–160 (2010).

    Article  PubMed  CAS  Google Scholar 

  13. Zheng, Y. et al. In vitro microvessels for the study of angiogenesis and thrombosis. Proc. Natl. Acad. Sci. USA 109, 9342–9347 (2012).

  14. Morgan, J. P. et al. Formation of microvascular networks in vitro. Nat. Protoc. 8, 1820 (2013).

  15. Hsieh, P. C. H., Davis, M. E., Lisowski, L. K. & Lee, R. T. Endothelial-cardiomyocyte interactions in cardiac development and repair. Annu. Rev. Physiol. 68, 51–66 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Lai, B. F. L. et al. InVADE: integrated vasculature for assessing dynamic events. Adv. Funct. Mater. 27, 1703524 (2017).

  17. Chiu, L. L., Montgomery, M., Liang, Y., Liu, H. & Radisic, M. Perfusable branching microvessel bed for vascularization of engineered tissues. Proc. Natl. Acad. Sci. USA 109, E3414–E3423 (2012).

  18. Bae, H. et al. Building vascular networks. Sci. Transl. Med. 4, 160ps123 (2012).

    Article  CAS  Google Scholar 

  19. Ren, X. et al. Engineering pulmonary vasculature in decellularized rat and human lungs. Nat. Biotechnol. 33, 1097–1102 (2015).

  20. Ott, H. C. et al. Perfusion-decellularized matrix: using nature’s platform to engineer a bioartificial heart. Nat. Med. 14, 213–221 (2008).

    Article  PubMed  CAS  Google Scholar 

  21. Ott, H. C. et al. Regeneration and orthotopic transplantation of a bioartificial lung. Nat. Med. 16, 927–933 (2010).

    Article  PubMed  CAS  Google Scholar 

  22. Kolesky, D. B. et al. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv. Mater. 26, 3124–3130 (2014).

    Article  PubMed  CAS  Google Scholar 

  23. Miller, J. S. et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat. Mater. 11, 768–774 (2012).

  24. Vollert, I. et al. In vitro perfusion of engineered heart tissue through endothelialized channels. Tissue Eng. Part A 20, 854–863 (2013).

  25. Tang, M. D., Golden, A. P. & Tien, J. Fabrication of collagen gels that contain patterned, micrometer-scale cavities. Adv. Mater. 16, 1345–1348 (2004).

    Article  CAS  Google Scholar 

  26. Golden, A. P. & Tien, J. Fabrication of microfluidic hydrogels using molded gelatin as a sacrificial element. Lab Chip 7, 720–725 (2007).

    Article  PubMed  CAS  Google Scholar 

  27. Zimmermann, W.-H. et al. Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts. Nat. Med. 12, 452–458 (2006).

    Article  PubMed  CAS  Google Scholar 

  28. Nunes, S. S. et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat. Methods 10, 781–787 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Leng, L., McAllister, A., Zhang, B., Radisic, M. & Günther, A. Mosaic hydrogels: one‐step formation of multiscale soft materials. Adv. Mater. 24, 3650–3658 (2012).

    Article  PubMed  CAS  Google Scholar 

  30. Kolesky, D. B. et al. 3D bioprinting of vascularized, heterogeneous cell‐laden tissue constructs. Adv. Mater. 26, 3124–3130 (2014).

    Article  PubMed  CAS  Google Scholar 

  31. Kolesky, D. B., Homan, K. A., Skylar-Scott, M. A. & Lewis, J. A. Three-dimensional bioprinting of thick vascularized tissues. Proc. Natl Acad. Sci. USA 113, 3179–3184 (2016).

  32. Kang, H.-W. et al. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nat. Biotechnol. 34, 312–319 (2016).

  33. Engelmayr, G. C. et al. Accordion-like honeycombs for tissue engineering of cardiac anisotropy. Nat. Mater. 7, 1003–1010 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Kolewe, M. E. et al. 3D structural patterns in scalable, elastomeric scaffolds guide engineered tissue architecture. Adv. Mater. 25, 4459–4465 (2013).

  35. Xiao, Y. et al. Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle. Lab Chip 14, 869–882 (2014).

  36. Zhang, B. et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nat. Mater. 15, 669–678 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Zhang, B., Montgomery, M., Davenport-Huyer, L., Korolj, A. & Radisic, M. Platform technology for scalable assembly of instantaneously functional mosaic tissues. Sci. Adv. 1, e1500423 (2015).

  38. Davenport Huyer, L. et al. Highly elastic and moldable polyester biomaterial for cardiac tissue engineering applications. ACS Biomater. Sci. Eng. 2, 780–788 (2016).

    Article  CAS  Google Scholar 

  39. Zhang, B., Peticone, C., Murthy, S. K. & Radisic, M. A standalone perfusion platform for drug testing and target validation in micro-vessel networks. Biomicrofluidics 7, 044125 (2013).

    Article  PubMed Central  CAS  Google Scholar 

  40. Sekine, H. et al. In vitro fabrication of functional three-dimensional tissues with perfusable blood vessels. Nat. Commun. 4, 1399 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Glaser, A. K. et al. Light-sheet microscopy for slide-free non-destructive pathology of large clinical specimens. Nat. Biomed. Eng. 1, 0084 (2017).

  42. Perbellini, F. et al. Free-of-acrylamide SDS-based tissue clearing (FASTClear) for three dimensional visualization of myocardial tissue. Sci. Rep. 7, 5188 (2017).

  43. Chung, K. & Deisseroth, K. Clarity for mapping the nervous system. Nat. Methods 10, 508–513 (2013).

    Article  PubMed  CAS  Google Scholar 

  44. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Coppeta, J. et al. A portable and reconfigurable multi-organ platform for drug development with onboard microfluidic flow control. Lab Chip 17, 134–144 (2017).

    Article  CAS  Google Scholar 

  46. Huh, D. et al. Reconstituting organ-level lung functions on a chip. Science 328, 1662–1668 (2010).

    Article  PubMed  CAS  Google Scholar 

  47. Shin, Y. et al. Microfluidic assay for simultaneous culture of multiple cell types on surfaces or within hydrogels. Nat. Protoc. 7, 1247–1259 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Tran, R. T. et al. Synthesis and characterization of a biodegradable elastomer featuring a dual crosslinking mechanism. Soft Matter. 6, 2449–2461 (2010).

  49. Yang, J., Webb, A. R. & Ameer, G. A. Novel citric acid-based biodegradable elastomers for tissue engineering. Adv. Mater. 16, 511–516 (2004).

    Article  CAS  Google Scholar 

  50. Montgomery, M. et al. Flexible shape-memory scaffold for minimally invasive delivery of functional tissues. Nat. Mater. 16, 1038–1046 (2017).

    Article  PubMed  CAS  Google Scholar 

  51. Khan, O. F., Voice, D. N., Leung, B. M. & Sefton, M. V. A novel high‐speed production process to create modular components for the bottom‐up assembly of large‐scale tissue‐engineered constructs. Adv. Healthc. Mater. 4, 113–120 (2015).

  52. Chamberlain, M. D., Gupta, R. & Sefton, M. V. Bone marrow-derived mesenchymal stromal cells enhance chimeric vessel development driven by endothelial cell-coated microtissues. Tissue Eng. A 18, 285–294 (2012).

    Article  CAS  Google Scholar 

  53. Radisic, M. et al. Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds. Proc. Natl. Acad. Sci. USA 101, 18129–18134 (2004).

  54. Zhang, B., Green, J. V., Murthy, S. K. & Radisic, M. Label-free enrichment of functional cardiomyocytes using microfluidic deterministic lateral flow displacement. PLoS ONE 7, e37619 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Ogawa, M. et al. Directed differentiation of cholangiocytes from human pluripotent stem cells. Nat. Biotechnol. 33, 853–861 (2015).

    Article  PubMed  CAS  Google Scholar 

  56. Ogawa, S. et al. Three-dimensional culture and cAMP signaling promote the maturation of human pluripotent stem cell-derived hepatocytes. Development 140, 3285–3296 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Lian, X. et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat. Protoc. 8, 162–175 (2013).

    Article  PubMed  CAS  Google Scholar 

  58. Levario, T. J., Zhan, M., Lim, B., Shvartsman, S. Y. & Lu, H. Microfluidic trap array for massively parallel imaging of Drosophila embryos. Nat. Protoc. 8, 721–736 (2013).

  59. Huh, D. et al. Microfabrication of human organs-on-chips. Nat. Protoc. 8, 2135–2157 (2013).

    Article  PubMed  CAS  Google Scholar 

  60. Kim, J. et al. A microfluidic technique for quantification of steroids in core needle biopsies. Anal. Chem. 87, 4688–4695 (2015).

    Article  PubMed  CAS  Google Scholar 

  61. Di, L. et al. Optimization of a higher throughput microsomal stability screening assay for profiling drug discovery candidates. J. Biomol. Screen. 8, 453–462 (2003).

    Article  PubMed  CAS  Google Scholar 

  62. Hiebert, L. M., Ping, T. & Wice, S. M. Repeated doses of oral and subcutaneous heparins have similar antithrombotic effects in a rat carotid arterial model of thrombosis. J. Cardiovasc. Pharmacol. Ther. 17, 110–116 (2012).

  63. Ross, M. H. & Pawlina, W. Histology (Lippincott Williams & Wilkins, Philadelphia, 2006).

  64. Zhang, K., Rekhter, M. D., Gordon, D. & Phan, S. H. Myofibroblasts and their role in lung collagen gene expression during pulmonary fibrosis. A combined immunohistochemical and in situ hybridization study. Am. J. Pathol. 145, 114 (1994).

    PubMed  PubMed Central  CAS  Google Scholar 

  65. JoVE Science Education Database. General laboratory techniques: histological sample preparation for light microscopy, https://www.jove.com/science-education/5039/histological-sample-preparation-for-light-microscopy (JoVE, Cambridge, MA, 2018).

Download references

Acknowledgements

This work was made possible by a National Sciences and Engineering Research Council of Canada (NSERC) Postgraduate Scholarship–Doctoral, awarded to B.F.L.L.; a Canadian Institutes of Health Research (CIHR) Vanier Canada Graduate Scholarship, awarded to L.D.H.; and a CIHR Banting Postdoctoral Fellowship, awarded to B.Z. This work was also funded by CIHR Operating Grants (MOP-126027 and MOP-137107), an NSERC Discovery Grant (RGPIN-2015-05952), an NSERC Steacie Fellowship (SMFSU 4620), a Heart and Stroke Foundation Grant-in-Aid (G-16-00012), an NSERC-CIHR Collaborative Health Research Grant (CHRPJ 4937), an NSERC Strategic Grant (STPGP 5066) to M.R., and National Institutes of Health grant 2R01 HL076485.

Author information

Authors and Affiliations

Authors

Contributions

B.Z. and M.R. designed the research; B.Z., L.D.H., and M.M. performed the research; B.Z. and M.R. analyzed the data; B.Z. and R.X. prepared the figures for the paper; B.Z., B.F.L.L., and R.X. prepared the supplementary video; B.Z., B.F.L.L., and M.R. wrote the manuscript.

Corresponding author

Correspondence to Milica Radisic.

Ethics declarations

Competing interests

M.R. and B.Z. are among the co-founders of TARA Biosystems and they hold equity in this company. The AngioChip is licensed to TARA Biosystems. The remaining 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.

Related links

Key references using this protocol

Zhang, B. et al. Nat. Mater. 15, 669–678 (2016) https://doi.org/10.1038/nmat4570

Lai., B. F. L. et al. Adv. Funct. Mater. 27, 1703524 (2017) https://doi.org/10.1002/adfm.201703524

Zhang, B. et al. Sci. Adv. 1, e1500423 (2015) https://doi.org/10.1126/sciadv.1500423

Integrated supplementary information

Supplementary Figure 1 Bioreactor design.

All units are in millimeters unless specified otherwise. All bioreactor components are made of polycarbonate.

Supplementary Figure 2 Endothelialized AngioChip network.

Confocal fluorescent images of various location of the AngioChip network populated with human umbilical cord endothelial cells stained for VE-cadherin (red) to identify the intercellular junctions (red) on day 2 after cell seeding. Scale bar, 50 μm. Reproduced with permission from Zhang et al.36, Macmillan Publishers Limited.

Supplementary Figure 3 Angiogenic vascular sprouting from AngioChip.

Confocal fluorescent images of GFP-labeled human umbilical cord endothelial cells (green) sprouting from the AngioChip networks through the built-in micro-holes on the channel walls in the presence of thymosin β4 on day 2 after cell seeding. Scale bar, 50μm.

Supplementary Figure 4 Tissue integration of AngioChip implants.

Smooth muscle actin (SMA) stained histology cross-section of the AngioChip cardiac tissue implants after 1 week with the direct surgical anastomosis in the configuration of artery-to-vein graft. Scale bar: 200μm (top image) and 100μm (bottom image). Reproduced with permission from Zhang et al.36, Macmillan Publishers Limited.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4

Supplementary Data

CAD file containing AngioChip design. Dimensions of four different AngioChips with increasing complexity. The default unit for dimensions in AutoCAD is the millimeter. The thickness of all layers is specified in micrometers

Supplementary Video 1

Step-by-step video protocol on the fabrication of AngioChip scaffolds. Step number in the video corresponds to the step number in the main protocol

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, B., Lai, B.F.L., Xie, R. et al. Microfabrication of AngioChip, a biodegradable polymer scaffold with microfluidic vasculature. Nat Protoc 13, 1793–1813 (2018). https://doi.org/10.1038/s41596-018-0015-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0015-8

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