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.

Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis

Abstract

We report the fabrication of a scaffold (hereafter referred to as AngioChip) that supports the assembly of parenchymal cells on a mechanically tunable matrix surrounding a perfusable, branched, three-dimensional microchannel network coated with endothelial cells. The design of AngioChip decouples the material choices for the engineered vessel network and for cell seeding in the parenchyma, enabling extensive remodelling while maintaining an open-vessel lumen. The incorporation of nanopores and micro-holes in the vessel walls enhances permeability, and permits intercellular crosstalk and extravasation of monocytes and endothelial cells on biomolecular stimulation. We also show that vascularized hepatic tissues and cardiac tissues engineered by using AngioChips process clinically relevant drugs delivered through the vasculature, and that millimetre-thick cardiac tissues can be engineered in a scalable manner. Moreover, we demonstrate that AngioChip cardiac tissues implanted with direct surgical anastomosis to the femoral vessels of rat hindlimbs establish immediate blood perfusion.

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: AngioChip scaffold fabrication and visualization.
Figure 2: Physical characterization of the AngioChip scaffolds.
Figure 3: Endothelialization of the AngioChip network.
Figure 4: Vascularized hepatic tissue assembly.
Figure 5: Vascularized cardiac tissue assembly.
Figure 6: Surgical anastomosis of the cardiac tissue.

References

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

    CAS  Google Scholar 

  2. Kim, H. J., Huh, D., Hamilton, G. & Ingber, D. E. Human gut-on-a-chip inhabited by microbial flora that experiences intestinal peristalsis-like motions and flow. Lab Chip 12, 2165–2174 (2012).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

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

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  6. Yang, X., Pabon, L. & Murry, C. E. Engineering adolescence maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Res. 114, 511–523 (2014).

    CAS  Google Scholar 

  7. Bian, W., Badie, N., Himel IV, H. D. & Bursac, N. Robust T-tubulation and maturation of cardiomyocytes using tissue-engineered epicardial mimetics. Biomaterials 35, 3819–3828 (2014).

    CAS  Google Scholar 

  8. Takebe, T. et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481–484 (2013).

    CAS  Google Scholar 

  9. Thavandiran, N. et al. Design and formulation of functional pluripotent stem cell-derived cardiac microtissues. Proc. Natl Acad. Sci. USA 110, E4698–E4707 (2013).

    CAS  Google Scholar 

  10. Legant, W. R. et al. Microfabricated tissue gauges to measure and manipulate forces from 3D microtissues. Proc. Natl Acad. Sci. USA 106, 10097–10102 (2009).

    CAS  Google Scholar 

  11. Bian, W. Engineered skeletal muscle tissue networks with controllable architecture. Biomaterials 30, 1401–1412 (2009).

    CAS  Google Scholar 

  12. Kim, S., Lee, H., Chung, M. & Jeon, N. L. Engineering of functional, perfusable 3D microvascular networks on a chip. Lab Chip 13, 1489–1500 (2013).

    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).

    CAS  Google Scholar 

  14. 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).

    Google Scholar 

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

    Google Scholar 

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

    Google Scholar 

  17. Ye, L., Zimmermann, W.-H., Garry, D. J. & Zhang, J. Patching the heart cardiac repair from within and outside. Circ. Res. 113, 922–932 (2013).

    CAS  Google Scholar 

  18. Baranski, J. D. et al. Geometric control of vascular networks to enhance engineered tissue integration and function. Proc. Natl Acad. Sci. USA 110, 7586–7591 (2013).

    CAS  Google Scholar 

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

    Google Scholar 

  20. Shandalov, Y. et al. An engineered muscle flap for reconstruction of large soft tissue defects. Proc. Natl Acad. Sci. USA 111, 6010–6015 (2014).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

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

    Google Scholar 

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

    CAS  Google Scholar 

  24. Ye, X. et al. A biodegradable microvessel scaffold as a framework to enable vascular support of engineered tissues. Biomaterials 34, 10007–10015 (2013).

    CAS  Google Scholar 

  25. Bettinger, C. J. J. et al. Three-dimensional microfluidic tissue-engineering scaffolds using a flexible biodegradable polymer. Adv. Mater. 18, 165–169 (2006).

    CAS  Google Scholar 

  26. Bettinger, C. J. et al. Silk fibroin microfluidic devices. Adv. Mater. 19, 2847–2850 (2007).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  29. Spiller, K., Freytes, D. & Vunjak-Novakovic, G. Macrophages modulate engineered human tissues for enhanced vascularization and healing. Ann. Biomed. Eng. 43, 616–627 (2014).

    Google Scholar 

  30. Kibbe, M. R. et al. Citric acid-based elastomers provide a biocompatible interface for vascular grafts. J. Biomed. Mater. Res. A 93A, 314–324 (2010).

    CAS  Google Scholar 

  31. Motlagh, D. et al. Hemocompatibility evaluation of poly(diol citrate) in vitro for vascular tissue engineering. J. Biomed. Mater. Res. A 82A, 907–916 (2007).

    CAS  Google Scholar 

  32. Dendukuri, D., Pregibon, D. C., Collins, J., Hatton, T. A. & Doyle, P. S. Continuous-flow lithography for high-throughput microparticle synthesis. Nature Mater. 5, 365–369 (2006).

    CAS  Google Scholar 

  33. Derby, B. Printing and prototyping of tissues and scaffolds. Science 338, 921–926 (2012).

    CAS  Google Scholar 

  34. Hoshi, R. A. Nanoporous biodegradable elastomers. Adv. Mater. 21, 188–192 (2009).

    CAS  Google Scholar 

  35. Nagueh, S. F. et al. Altered titin expression, myocardial stiffness, and left ventricular function in patients with dilated cardiomyopathy. Circulation 110, 155–162 (2004).

    CAS  Google Scholar 

  36. Weis, S. M. et al. Myocardial mechanics and collagen structure in the osteogenesis imperfecta murine (oim). Circ. Res. 87, 663–669 (2000).

    CAS  Google Scholar 

  37. Coirault, C. et al. Increased compliance in diaphragm muscle of the cardiomyopathic Syrian hamster. J. Appl. Physiol. 85, 1762–1769 (1998).

    CAS  Google Scholar 

  38. Omens, J. H. Stress and strain as regulators of myocardial growth. Prog. Biophys. Mol. Biol. 69, 559–572 (1998).

    CAS  Google Scholar 

  39. Yeh, W. C. et al. Elastic modulus measurements of human liver and correlation with pathology. Ultrasound Med. Biol. 28, 467–474 (2002).

    Google Scholar 

  40. Merkel, T. C., Bondar, V. I., Nagai, K., Freeman, B. D. & Pinnau, I. Gas sorption, diffusion, and permeation in poly(dimethylsiloxane). J. Polym. Sci. B 38, 415–434 (2000).

    CAS  Google Scholar 

  41. Toepke, M. W. & Beebe, D. J. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip 6, 1484–1486 (2006).

    CAS  Google Scholar 

  42. Gaillard, P. J. et al. Establishment and functional characterization of an in vitro model of the blood-brain barrier, comprising a co-culture of brain capillary endothelial cells and astrocytes. Eur. J. Pharm. Sci. 12, 215–222 (2001).

    CAS  Google Scholar 

  43. Yuan, W., Lv, Y., Zeng, M. & Fu, B. M. Non-invasive measurement of solute permeability in cerebral microvessels of the rat. Microvasc. Res. 77, 166–173 (2009).

    CAS  Google Scholar 

  44. Adamson, R. H., Huxley, V. H. & Curry, F. E. Single capillary permeability to proteins having similar size but different charge. Am. J. Physiol. 254, H304–H312 (1988).

    CAS  Google Scholar 

  45. Woosley, R. L., Chen, Y., Freiman, J. P. & Gillis, R. A. Mechanism of the cardiotoxic actions of terfenadine. JAMA 269, 1532–1536 (1993).

    CAS  Google Scholar 

  46. 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).

    CAS  Google Scholar 

  47. Boudou, T. et al. A microfabricated platform to measure and manipulate the mechanics of engineered cardiac microtissues. Tissue Eng. A 18, 910–919 (2012).

    CAS  Google Scholar 

  48. 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).

    CAS  Google Scholar 

  49. Tulloch, N. L. et al. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ. Res. 109, 47–59 (2011).

    CAS  Google Scholar 

  50. Wu, W., Allen, R. A. & Wang, Y. Fast-degrading elastomer enables rapid remodeling of a cell-free synthetic graft into a neoartery. Nature Med. 18, 1148–1153 (2012).

    CAS  Google Scholar 

  51. Bhatia, S., Balis, U., Yarmush, M. & Toner, M. Effect of cell–cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J. 13, 1883–1900 (1999).

    CAS  Google Scholar 

  52. Mazza, E. & Ehret, A. E. Mechanical biocompatibility of highly deformable biomedical materials. J. Mech. Behav. Biomed. Mater. 48, 100–124 (2015).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  54. Nava, A., Mazza, E., Furrer, M., Villiger, P. & Reinhart, W. In vivo mechanical characterization of human liver. Med. Image Anal. 12, 203–216 (2008).

    CAS  Google Scholar 

  55. Hoshi, R. A. et al. The blood and vascular cell compatibility of heparin-modified ePTFE vascular grafts. Biomaterials 34, 30–41 (2013).

    CAS  Google Scholar 

  56. Sefton, M. V., Gemmell, C. H. & Gorbet, M. B. in Biomaterials Science 3rd edn (eds Ratner, B. D., Hoffman, A. S., Schoen, F. J. & Lemons, J. E.) 758–760 (Academic, 2013).

    Google Scholar 

  57. 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).

    CAS  Google Scholar 

  58. Ogawa, M. et al. Directed differentiation of cholangiocytes from human pluripotent stem cells. Nature Biotech. 33, 853–861 (2015).

    CAS  Google Scholar 

  59. Berry, M. & Friend, D. High-yield preparation of isolated rat liver parenchymal cells: a biochemical and fine structural study. J. Cell Biol. 43, 506–520 (1969).

    CAS  Google Scholar 

  60. Kennedy, M., D’Souza, S. L., Lynch-Kattman, M., Schwantz, S. & Keller, G. Development of the hemangioblast defines the onset of hematopoiesis in human ES cell differentiation cultures. Blood 109, 2679–2687 (2007).

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  62. Zeebregts, C., Heijmen, R., Van Den Dungen, J. & Van Schilfgaarde, R. Non-methods of vascular anastomosis. Br. J. Surg. 90, 261–271 (2003).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Marjan and P. Lai from the University Health Network, Toronto, for their help in the optical mapping analysis. We thank Y. Liu from Osaka University, Japan, for her help in quantifying the platelet coverage on the AngioChip in the blood perfusion study. We thank J. W. Miklas and Y. Xiao for their helpful discussion regarding human cardiomyocyte culture and cell seeding. We thank A. Sofla for his help with the POMaC synthesis. We thank A. Keating and I. Rashedi for providing hMSCs and Y. Zhao for her help in culturing and expanding hMSCs. We thank J. Yang for suggestions regarding POMaC synthesis. This work was made possible by the National Sciences and Engineering Research Council of Canada (NSERC) Steacie Fellowship to M.R. This work was also financially supported by the Canadian Institutes of Health Research (CIHR) Operating Grants (MOP-126027 and MOP-137107), the Heart and Stroke Foundation GIA T6946, NSERC–CIHR Collaborative Health Research Grant (CHRPJ 385981-10), NSERC Discovery Grant (RGPIN 326982-10), NSERC Discovery Accelerator Supplement (RGPAS 396125-10) and National Institutes of Health Grant 2R01 HL076485.

Author information

Authors and Affiliations

Authors

Contributions

B.Z. developed the AngioChip concept, designed and performed experiments, analysed data and prepared the manuscript. M.M. contributed to mechanical testing, polymer characterization, sprouting assay, blood perfusion experiments, and vascular anastomosis surgery. M.D.C. performed the primary rat hepatocyte isolation and urea assay. S.O. differentiated hESC-derived hepatocytes. A.K. performed polymer mechanical testing. A.P. differentiated hESC-derived cardiomyocytes and contributed to the whole blood perfusion experiment and optical mapping. L.A.W. performed extraction of human whole blood. S.M. and K.N. performed optical mapping measurements and analysis. J.K. performed mass spectrometry analysis. L.R. contributed to the direct vascular anastomosis surgery; A.M. performed the direct vascular anastomosis surgery; S.S.N. contributed to the direct vascular anastomosis surgery and writing of the manuscript. A.R.W. contributed to the writing of the manuscript. G.K. contributed to the writing of the manuscript. M.V.S. contributed to writing of the manuscript. M.R. developed the AngioChip concept, supervised the work and wrote the manuscript.

Corresponding author

Correspondence to Milica Radisic.

Ethics declarations

Competing interests

M.R. and B.Z. are amongst co-founders of TARA Biosystems and they hold equity in this company.

Supplementary information

Supplementary Information

Supplementary Information (PDF 25074 kb)

Supplementary Movie 1

Supplementary Movie 1 (MOV 4710 kb)

Supplementary Movie 2

Supplementary Movie 2 (MOV 2917 kb)

Supplementary Movie 3

Supplementary Movie 3 (MOV 4817 kb)

Supplementary Movie 4

Supplementary Movie 4 (MOV 8083 kb)

Supplementary Movie 5

Supplementary Movie 5 (MOV 6213 kb)

Supplementary Movie 6

Supplementary Movie 6 (MOV 511 kb)

Supplementary Movie 7

Supplementary Movie 7 (MOV 9570 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, B., Montgomery, M., Chamberlain, M. et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nature Mater 15, 669–678 (2016). https://doi.org/10.1038/nmat4570

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmat4570

This article is cited by

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