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:

Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing

Abstract

Biomedical research has relied on animal studies and conventional cell cultures for decades. Recently, microphysiological systems (MPS), also known as organs-on-chips, that recapitulate the structure and function of native tissues in vitro, have emerged as a promising alternative1. However, current MPS typically lack integrated sensors and their fabrication requires multi-step lithographic processes2. Here, we introduce a facile route for fabricating a new class of instrumented cardiac microphysiological devices via multimaterial three-dimensional (3D) printing. Specifically, we designed six functional inks, based on piezo-resistive, high-conductance, and biocompatible soft materials that enable integration of soft strain gauge sensors within micro-architectures that guide the self-assembly of physio-mimetic laminar cardiac tissues. We validated that these embedded sensors provide non-invasive, electronic readouts of tissue contractile stresses inside cell incubator environments. We further applied these devices to study drug responses, as well as the contractile development of human stem cell-derived laminar cardiac tissues over four weeks.

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: Device principle and microscale 3D-printing procedure.
Figure 2: Micro-grooves guide cardiomyocyte self-assembly into anisotropic engineered tissues.
Figure 3: CB:TPU gauge factor, sensor readout and example drug-dose studies.
Figure 4: Long-term hips-CM contractile development and thicker laminar NRVM tissue devices.

Similar content being viewed by others

References

  1. Bhatia, S. N. & Ingber, D. E. Microfluidic organs-on-chips. Nat. Biotechnol. 32, 760–772 (2014).

    Article  CAS  Google Scholar 

  2. Mammoto, A. et al. Control of lung vascular permeability and endotoxin-induced pulmonary oedema by changes in extracellular matrix mechanics. Nat. Commun. 4, 1759 (2013).

    Article  Google Scholar 

  3. Feinberg, A. W. et al. Muscular thin films for building actuators and powering devices. Science 317, 1366–1370 (2007).

    Article  CAS  Google Scholar 

  4. Boudou, T. et al. A microfabricated platform to measure and manipulate the mechanics of engineered cardiac microtissues. Tissue Eng. A 18, 910–919 (2011).

    Article  Google Scholar 

  5. Wang, G. et al. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat. Med. 20, 616–623 (2014).

    Article  CAS  Google Scholar 

  6. Hinson, J. T. et al. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349, 982–986 (2015).

    Article  CAS  Google Scholar 

  7. Agarwal, A. et al. Micropatterning alginate substrates for in vitro cardiovascular muscle on a chip. Adv. Funct. Mater. 23, 3738–3746 (2013).

    Article  CAS  Google Scholar 

  8. Grosberg, A., Alford, P. W., McCain, M. L. & Parker, K. K. Ensembles of engineered cardiac tissues for physiological and pharmacological study: heart on a chip. Lab Chip 11, 4165–4173 (2011).

    Article  CAS  Google Scholar 

  9. Park, S.-J. et al. Phototactic guidance of a tissue-engineered soft-robotic ray. Science 353, 158–162 (2016).

    Article  CAS  Google Scholar 

  10. Lewis, J. A. Direct ink writing of 3D functional materials. Adv. Funct. Mater. 16, 2193–2204 (2006).

    Article  CAS  Google Scholar 

  11. Hardin, J. O., Ober, T. J., Valentine, A. D. & Lewis, J. A. Microfluidic printheads for multimaterial 3D printing of viscoelastic inks. Adv. Mater. 27, 3279–3284 (2015).

    Article  CAS  Google Scholar 

  12. Sun, K. et al. 3D printing of interdigitated Li-Ion microbattery architectures. Adv. Mater. 25, 4539–4543 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  14. 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).

    Article  CAS  Google Scholar 

  15. Matsuhisa, N. et al. Printable elastic conductors with a high conductivity for electronic textile applications. Nat. Commun. 6, 7461 (2015).

    Article  CAS  Google Scholar 

  16. Lipomi, D. J. et al. Skin-like pressure and strain sensors based on transparent elastic films of carbon nanotubes. Nat. Nanotech. 6, 788–792 (2011).

    Article  CAS  Google Scholar 

  17. Berthier, E., Young, E. W. & Beebe, D. Engineers are from PDMS-land, biologists are from polystyrenia. Lab Chip 12, 1224–1237 (2012).

    Article  CAS  Google Scholar 

  18. Kléber, A. G. & Rudy, Y. Basic mechanisms of cardiac impulse propagation and associated arrhythmias. Physiol. Rev. 84, 431–488 (2004).

    Article  Google Scholar 

  19. Pasqualini, F. S., Sheehy, S. P., Agarwal, A., Aratyn-Schaus, Y. & Parker, K. K. Structural phenotyping of stem cell-derived cardiomyocytes. Stem Cell Rep. 4, 340–347 (2015).

    Article  CAS  Google Scholar 

  20. Feinberg, A. W. et al. Controlling the contractile strength of engineered cardiac muscle by hierarchal tissue architecture. Biomaterials 33, 5732–5741 (2012).

    Article  CAS  Google Scholar 

  21. Sheehy, S. P. et al. Quality metrics for stem cell-derived cardiac myocytes. Stem Cell Rep. 2, 282–294 (2014).

    Article  CAS  Google Scholar 

  22. OštIádalová, I. et al. Early postnatal development of contractile performance and responsiveness to Ca2+, verapamil and ryanodine in the isolated rat heart. J. Mol. Cell. Cardiol. 25, 733–740 (1993).

    Article  Google Scholar 

  23. Zhang, D. et al. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 34, 5813–5820 (2013).

    Article  CAS  Google Scholar 

  24. Zimmermann, W. H. et al. Three-dimensional engineered heart tissue from neonatal rat cardiac myocytes. Biotechnol. Bioeng. 68, 106–114 (2000).

    Article  CAS  Google Scholar 

  25. Reiser, P. J., Portman, M. A., Ning, X.-H. & Moravec, C. S. Human cardiac myosin heavy chain isoforms in fetal and failing adult atria and ventricles. Am. J. Physiol. 280, H1814–H1820 (2001).

    CAS  Google Scholar 

  26. Lundy, S. D., Zhu, W-Z., Regnier, M. & Laflamme, M. A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 22, 1991–2002 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  29. Zimmermann, W.-H. et al. Tissue engineering of a differentiated cardiac muscle construct. Circ. Res. 90, 223–230 (2002).

    Article  CAS  Google Scholar 

  30. LeGrice, I. J. et al. Laminar structure of the heart: ventricular myocyte arrangement and connective tissue architecture in the dog. Am. J. Physiol. 269, H571–H582 (1995).

    CAS  Google Scholar 

Download references

Acknowledgements

The authors thank L. K. Sanders for her work on photography and time-lapse movies, J. A. Goss for his assistance with fabrication of the device holder and J. Minardi for his development of Mecode, and his help with machine automation. This work was performed in part at the Center for Nanoscale Systems (CNS), a member of the National Nanotechnology Infrastructure Network (NNIN), which is supported by the National Science Foundation under NSF award no. ECS-0335765. CNS is part of Harvard University. This work was also supported by the National Center For Advancing Translational Sciences of the National Institutes of Health under Award Number UH3TR000522, the US Army Research Laboratory and the US Army Research Office under Contract No. W911NF-12-2-0036, the Air Force Research Laboratory under Contract No. FA8650-09-D-5037-0004, and the Harvard University Materials Research Science and Engineering Center (MRSEC) award no. DMR-1420570. J.U.L. gratefully acknowledges support from the Villum Foundation. J.A.L. gratefully acknowledges support from the Office of Naval Research, Vannevar Bush National Security Science and Engineering Faculty Fellowship (Award No. N00014-16-1-2823).

Author information

Authors and Affiliations

Authors

Contributions

J.U.L., T.A.B., J.A.L. and K.K.P. designed the study. J.U.L. and T.A.B. designed the device. T.A.B. coded the 3D-print procedure and automation. J.U.L., T.A.B., A.K. and A.D.V. developed and characterized the printable materials. J.U.L., A.D.V. and T.A.B., optimized and printed devices. P.H.C. performed NRVM harvesting and prepared culturing media. J.U.L., M.Y. and A.P.N. performed NRVM culture, drug-dose experiments, and data analysis. M.Y. and J.U.L. conducted hiPS-CM culture, experiments and data analysis. F.S.P. and J.U.L. performed tissue staining, confocal imaging, and OOP analysis. S.-J.P. and J.U.L. conducted optical mapping experiments and analysis. H.Y. and J.J.V. developed the mechanical model of the device. J.U.L., T.A.B., J.A.L. and K.K.P. prepared illustrations and wrote the manuscript. F.S.P. and A.D.V. contributed to writing the manuscript.

Corresponding authors

Correspondence to Jennifer A. Lewis or Kevin K. Parker.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 5849 kb)

Supplementary Information

Supplementary movie 1 (MOV 8624 kb)

Supplementary Information

Supplementary movie 2 (MOV 4801 kb)

Supplementary Information

Supplementary movie 3 (MOV 8813 kb)

Supplementary Information

Supplementary movie 4 (MOV 164 kb)

Supplementary Information

Supplementary movie 5 (MOV 151 kb)

Supplementary Information

Supplementary movie 6 (MOV 104 kb)

Supplementary Information

Supplementary movie 7 (MOV 148 kb)

Supplementary Information

Supplementary movie 8 (MOV 9398 kb)

Supplementary Information

Supplementary movie 9 (MOV 1207 kb)

Supplementary Information

Supplementary movie 10 (MOV 2084 kb)

Supplementary Information

Supplementary movie 11 (MOV 1382 kb)

Supplementary Information

Supplementary movie 12 (MOV 1674 kb)

Supplementary Information

Supplementary movie 13 (MOV 1631 kb)

Supplementary Information

Supplementary movie 14 (MOV 638 kb)

Supplementary Information

Supplementary movie 15 (MOV 2341 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lind, J., Busbee, T., Valentine, A. et al. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nature Mater 16, 303–308 (2017). https://doi.org/10.1038/nmat4782

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmat4782

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