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.

  • Perspective
  • Published:

Microfluidic organs-on-chips

Abstract

An organ-on-a-chip is a microfluidic cell culture device created with microchip manufacturing methods that contains continuously perfused chambers inhabited by living cells arranged to simulate tissue- and organ-level physiology. By recapitulating the multicellular architectures, tissue-tissue interfaces, physicochemical microenvironments and vascular perfusion of the body, these devices produce levels of tissue and organ functionality not possible with conventional 2D or 3D culture systems. They also enable high-resolution, real-time imaging and in vitro analysis of biochemical, genetic and metabolic activities of living cells in a functional tissue and organ context. This technology has great potential to advance the study of tissue development, organ physiology and disease etiology. In the context of drug discovery and development, it should be especially valuable for the study of molecular mechanisms of action, prioritization of lead candidates, toxicity testing and biomarker identification.

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: Fabrication methods for microfluidic chips.

Kim Caesar/Nature Publishing Group

Figure 2: Examples of increasingly complex single-channel, organ-on-chip designs.

Katie Vicari/Nature Publishing Group

Figure 3: Microfluidic chip models of angiogenesis and immune cell invasion that incorporate ECM gels.
Figure 4: Examples of increasingly complex organ-on-chip designs.
Figure 5: A multi-organ microfluidic framework used for PK/PD modeling.

Similar content being viewed by others

References

  1. Harrison, R.G. The outgrowth of the nerve fiber as a mode of protoplasmic movement. J. Exp. Zool. 9, 787–846 (1910).

    Article  Google Scholar 

  2. Greek, R. & Menache, A. Systematic reviews of animal models: methodology versus epistemology. Int. J. Med. Sci. 10, 206–221 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Ehrmann, R.L. & Gey, G.O. The growth of cells on a transparent gel of reconstituted rat-tail collagen. J. Natl. Cancer Inst. 16, 1375–1403 (1956).

    CAS  PubMed  Google Scholar 

  4. Mroue, R. & Bissell, M.J. Three-dimensional cultures of mouse mammary epithelial cells. Methods Mol. Biol. 945, 221–250 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Sato, T. & Clevers, H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanisms and applications. Science 340, 1190–1194 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Lancaster, M.A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Muranen, T. et al. Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. Cancer Cell 21, 227–239 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Mammoto, T., Mammoto, A. & Ingber, D.E. Mechanobiology and developmental control. Annu. Rev. Cell Dev. Biol. 29, 27–61 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Ingber, D.E. Mechanobiology and diseases of mechanotransduction. Ann. Med. 35, 564–577 (2003).

    Article  PubMed  Google Scholar 

  10. Duffy, D.C., McDonald, J.C., Schueller, O.J. & Whitesides, G.M. Rapid prototyping of microfluidic systems in poly(dimethylsiloxane). Anal. Chem. 70, 4974–4984 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Singhvi, R. et al. Engineering cell shape and function. Science 264, 696–698 (1994).

    Article  CAS  PubMed  Google Scholar 

  12. Chen, C.S., Mrksich, M., Huang, S., Whitesides, G. & Ingber, D.E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Folch, A. & Toner, M. Cellular micropatterns on biocompatible materials. Biotechnol. Prog. 14, 388–392 (1998).

    Article  CAS  PubMed  Google Scholar 

  14. Kane, R.S., Takayama, S., Ostuni, E., Ingber, D.E. & Whitesides, G.M. Patterning proteins and cells using soft lithography. Biomaterials 20, 2363–2376 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Folch, A., Ayon, A., Hurtado, O., Schmidt, M.A. & Toner, M. Molding of deep polydimethylsiloxane microstructures for microfluidics and biological applications. J. Biomech. Eng. 121, 28–34 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Douville, N.J. et al. Fabrication of two-layered channel system with embedded electrodes to measure resistance across epithelial and endothelial barriers. Anal. Chem. 82, 2505–2511 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nguyen, T.A., Yin, T.I., Reyes, D. & Urban, G.A. Microfluidic chip with integrated electrical cell-impedance sensing for monitoring single cancer cell migration in three-dimensional matrixes. Anal. Chem. 85, 11068–11076 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Liu, M.C. et al. Electrofluidic pressure sensor embedded microfluidic device: a study of endothelial cells under hydrostatic pressure and shear stress combinations. Lab Chip 13, 1743–1753 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Eklund, S.E. et al. Metabolic discrimination of select list agents by monitoring cellular responses in a multianalyte microphysiometer. Sensors (Basel) 9, 2117–2133 (2009).

    Article  CAS  Google Scholar 

  20. Takayama, S. et al. Subcellular positioning of small molecules. Nature 411, 1016 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Li Jeon, N. et al. Neutrophil chemotaxis in linear and complex gradients of interleukin-8 formed in a microfabricated device. Nat. Biotechnol. 20, 826–830 (2002).

    Article  PubMed  CAS  Google Scholar 

  22. Prentice-Mott, H.V. et al. Biased migration of confined neutrophil-like cells in asymmetric hydraulic environments. Proc. Natl. Acad. Sci. USA 110, 21006–21011 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Radisic, M., Deen, W.M., Langer, R. & Vunjak-Novakovic, G. Mathematical model of oxygen distribution in engineered cardiac tissue with parallel channel array perfused with culture medium containing oxygen carriers. Am. J. Physiol.Heart Care Physiol. 288, H1278–H1289 (2005).

    Article  CAS  Google Scholar 

  24. Xiao, R.R. et al. Simultaneous generation of gradients with gradually changed slope in a microfluidic device for quantifying axon response. Anal. Chem. 85, 7842–7850 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Peng, C.C., Liao, W.H., Chen, Y.H., Wu, C.Y. & Tung, Y.C. A microfluidic cell culture array with various oxygen tensions. Lab Chip 13, 3239–3245 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Cimetta, E. et al. Microfluidic device generating stable concentration gradients for long term cell culture: application to Wnt3a regulation of betacatenin signaling. Lab Chip 10, 3277–3283 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Seidi, A. et al. A microfluidic-based neurotoxin concentration gradient for the generation of an in vitro model of Parkinson's disease. Biomicrofluidics 5, 22214 (2011).

    Article  PubMed  CAS  Google Scholar 

  28. Chen, S. & Lee, L.P. Non-invasive microfluidic gap junction assay. Integr. Biol. (Camb) 2, 130–138 (2010).

    Article  CAS  Google Scholar 

  29. Carraro, A. et al. In vitro analysis of a hepatic device with intrinsic microvascular-based channels. Biomed. Microdevices 10, 795–805 (2008).

    Article  PubMed  Google Scholar 

  30. Griep, L.M. et al. BBB on chip: microfluidic platform to mechanically and biochemically modulate blood-brain barrier function. Biomed. Microdevices 15, 145–150 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Lee, P.J., Hung, P.J. & Lee, L.P. An artificial liver sinusoid with a microfluidic endothelial-like barrier for primary hepatocyte culture. Biotechnol. Bioeng. 97, 1340–1346 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Chiu, D.T. et al. Patterned deposition of cells and proteins onto surfaces by using three dimensional microfluidic systems. Proc. Natl. Acad. Sci. USA 97, 2408–2413 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kane, B.J., Zinner, M.J., Yarmush, M.L. & Toner, M. Liver-specific functional studies in a microfluidic array of primary mammalian hepatocytes. Anal. Chem. 78, 4291–4298 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. Tumarkin, E. et al. High-throughput combinatorial cell co-culture using microfluidics. Integr. Biol. (Camb) 3, 653–662 (2011).

    Article  CAS  Google Scholar 

  35. Agarwal, A., Goss, J.A., Cho, A., McCain, M.L. & Parker, K.K. Microfluidic heart on a chip for higher throughput pharmacological studies. Lab Chip 13, 3599–3608 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Esch, M.B. et al. On chip porous polymer membranes for integration of gastrointestinal tract epithelium with microfluidic 'body-on-a-chip' devices. Biomed. Microdevices 14, 895–906 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Baker, B.M., Trappmann, B., Stapleton, S.C., Toro, E. & Chen, C.S. Microfluidics embedded within extracellular matrix to define vascular architectures and pattern diffusive gradients. Lab Chip 13, 3246–3252 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nguyen, D.H. et al. Biomimetic model to reconstitute angiogenic sprouting morphogenesis in vitro. Proc. Natl. Acad. Sci. USA 110, 6712–6717 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Han, S. et al. A versatile assay for monitoring in vivo-like transendothelial migration of neutrophils. Lab Chip 12, 3861–3865 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Khanal, G., Chung, K., Solis-Wever, X., Johnson, B. & Pappas, D. Ischemia/reperfusion injury of primary porcine cardiomyocytes in a lowshear microfluidic culture and analysis device. Analyst (Lond.) 136, 3519–3526 (2011).

    Article  CAS  Google Scholar 

  41. Tsantoulas, C. et al. Probing functional properties of nociceptive axons using a microfluidic culture system. PLoS ONE 8, e80722 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Li, C.Y., Wood, D.K., Huang, J.H. & Bhatia, S.N. Flow-based pipeline for systematic modulation and analysis of 3D tumor microenvironments. Lab Chip 13, 1969–1978 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Viravaidya, K. & Shuler, M.L. Incorporation of 3T3–L1 cells to mimic bioaccumulation in a microscale cell culture analog device for toxicity studies. Biotechnol. Prog. 20, 590–597 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Huh, D. et al. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Sci. Transl. Med. 4, 159ra147 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jang, K.J. & Suh, K.Y. A multi-layer microfluidic device for efficient culture and analysis of renal tubular cells. Lab Chip 10, 36–42 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Booth, R. & Kim, H. Characterization of a microfluidic in vitro model of the blood-brain barrier (mBBB). Lab Chip 12, 1784–1792 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. 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).

    Article  CAS  PubMed  Google Scholar 

  49. Kim, H.J. & Ingber, D.E. Gut-on-a-Chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr Biol (Camb) 5, 1130–1140 (2013).

    Article  CAS  Google Scholar 

  50. Park, S.H. et al. Chip-based comparison of the osteogenesis of human bone marrow- and adipose tissue-derived mesenchymal stem cells under mechanical stimulation. PLoS ONE 7, e46689 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sun, Y.S., Peng, S.W. & Cheng, J.Y. In vitro electrical-stimulated wound-healing chip for studying electric field-assisted wound healing process. Biomicrofluidics 6, 34117 (2012).

    Article  PubMed  CAS  Google Scholar 

  52. Wan, C.R., Chung, S. & Kamm, R.D. Differentiation of embryonic stem cells into cardiomyocytes in a compliant microfluidic system. Ann. Biomed. Eng. 39, 1840–1847 (2011).

    Article  PubMed  Google Scholar 

  53. Feinberg, A.W. et al. Functional differences in engineered myocardium from embryonic stem cell-derived versus neonatal cardiomyocytes. Stem Cell Reports 1, 387–396 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cimetta, E. et al. Microfluidic bioreactor for dynamic regulation of early mesodermal commitment in human pluripotent stem cells. Lab Chip 13, 355–364 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Chung, B.G. et al. Human neural stem cell growth and differentiation in a gradient generating microfluidic device. Lab Chip 5, 401–406 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Yang, K. et al. A microfluidic array for quantitative analysis of human neural stem cell self-renewal and differentiation in three-dimensional hypoxic microenvironment. Biomaterials 34, 6607–6614 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Trkov, S., Eng, G., Di Liddo, R., Parnigotto, P.P. & Vunjak-Novakovic, G. Micropatterned three-dimensional hydrogel system to study human endothelial-mesenchymal stem cell interactions. J. Tissue Eng. Regen. Med. 4, 205–215 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Han, S. et al. Three-dimensional extracellular matrix-mediated neural stem cell differentiation in a microfluidic device. Lab Chip 12, 2305–2308 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Kroon, E. et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452 (2008).

    Article  CAS  PubMed  Google Scholar 

  60. Später, D. et al. A HCN4+ cardiomyogenic progenitor derived from the first heart field and human pluripotent stem cells. Nat. Cell Biol. 15, 1098–1106 (2013).

    Article  PubMed  CAS  Google Scholar 

  61. van Laake, L.W. et al. Reporter-based isolation of induced pluripotent stem cell- and embryonic stem cell-derived cardiac progenitors reveals limited gene expression variance. Circ. Res. 107, 340–347 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Shan, J. et al. High-throughput identification of small molecules for human hepatocyte expansion and iPS differentiation. Nat. Chem. Biol. 9, 514–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sin, A. et al. The design and fabrication of three-chamber microscale cell culture analog devices with integrated dissolved oxygen sensors. Biotechnol. Prog. 20, 338–345 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Chao, P., Maguire, T., Novik, E., Cheng, K.C. & Yarmush, M.L. Evaluation of a microfluidic based cell culture platform with primary human hepatocytes for the prediction of hepatic clearance in human. Biochem. Pharmacol. 78, 625–632 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Legendre, A. et al. Metabolic characterization of primary rat hepatocytes cultivated in parallel microfluidic biochips. J. Pharm. Sci. 102, 3264–3276 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Cheng, S., Prot, J.M., Leclerc, E. & Bois, F.Y. Zonation related function and ubiquitination regulation in human hepatocellular carcinoma cells in dynamic vs. static culture conditions. BMC Genomics 13, 54 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Allen, J.W. & Bhatia, S.N. Formation of steady-state oxygen gradients in vitro: application to liver zonation. Biotechnol. Bioeng. 82, 253–262 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Sivaraman, A. et al. A microscale in vitro physiological model of the liver: predictive screens for drug metabolism and enzyme induction. Curr. Drug Metab. 6, 569–591 (2005).

    Article  CAS  PubMed  Google Scholar 

  69. Toh, Y.C. et al. A microfluidic 3D hepatocyte chip for drug toxicity testing. Lab Chip 9, 2026–2035 (2009).

    Article  CAS  PubMed  Google Scholar 

  70. Novik, E., Maguire, T.J., Chao, P., Cheng, K.C. & Yarmush, M.L. A microfluidic hepatic coculture platform for cell-based drug metabolism studies. Biochem. Pharmacol. 79, 1036–1044 (2010).

    Article  CAS  PubMed  Google Scholar 

  71. Baudoin, R., Griscom, L., Monge, M., Legallais, C. & Leclerc, E. Development of a renal microchip for in vitro distal tubule models. Biotechnol. Prog. 23, 1245–1253 (2007).

    CAS  PubMed  Google Scholar 

  72. Snouber, L.C. et al. Analysis of transcriptomic and proteomic profiles demonstrates improved Madin-Darby canine kidney cell function in a renal microfluidic biochip. Biotechnol. Prog. 28, 474–484 (2012).

    Article  CAS  PubMed  Google Scholar 

  73. Jang, K.J. et al. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr. Biol. (Camb) 5, 1119–1129 (2013).

    Article  CAS  Google Scholar 

  74. Mahler, G.J., Esch, M.B., Glahn, R.P. & Shuler, M.L. Characterization of a gastrointestinal tract microscale cell culture analog used to predict drug toxicity. Biotechnol. Bioeng. 104, 193–205 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Huh, D. et al. Acoustically detectable cellular-level lung injury induced by fluid mechanical stresses in microfluidic airway systems. Proc. Natl. Acad. Sci. USA 104, 18886–18891 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Fritsche, C.S. et al. Pulmonary tissue engineering using dual-compartment polymer scaffolds with integrated vascular tree. Int. J. Artif. Organs. 32, 701–710 (2009).

    Article  CAS  PubMed  Google Scholar 

  77. Tavana, H. et al. Epithelium damage and protection during reopening of occluded airways in a physiologic microfluidic pulmonary airway model. Biomed. Microdevices 13, 731–742 (2011).

    Article  PubMed  Google Scholar 

  78. 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  PubMed  PubMed Central  Google Scholar 

  79. Cheng, W., Klauke, N., Sedgwick, H., Smith, G.L. & Cooper, J.M. Metabolic monitoring of the electrically stimulated single heart cell within a microfluidic platform. Lab Chip 6, 1424–1431 (2006).

    Article  CAS  PubMed  Google Scholar 

  80. Giridharan, G.A. et al. Microfluidic cardiac cell culture model (mCCCM). Anal. Chem. 82, 7581–7587 (2010).

    Article  CAS  PubMed  Google Scholar 

  81. Grosberg, A. et al. Muscle on a chip: in vitro contractility assays for smooth and striated muscle. J. Pharmacol. Toxicol. Methods 65, 126–135 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhang, Y., Gazit, Z., Pelled, G., Gazit, D. & Vunjak-Novakovic, G. Patterning osteogenesis by inducible gene expression in microfluidic culture systems. Integr. Biol. (Camb) 3, 39–47 (2011).

    Article  CAS  Google Scholar 

  83. Zhang, W., Lee, W.Y., Siegel, D.S., Tolias, P. & Zilberberg, J. Patient-specific 3D microfluidic tissue model for multiple myeloma. Tissue Eng. Part C Methods doi:10.1089/ten.tec.2013.0490 (17 January 2014).

  84. Torisawa, Y.S. et al. Bone marrow-on-a-chip replicates hematopoietic niche physiology in vitro. Nat. Methods 11, 663–669 (2014).

    Article  CAS  PubMed  Google Scholar 

  85. Puleo, C.M., McIntosh Ambrose, W., Takezawa, T., Elisseeff, J. & Wang, T.H. Integration and application of vitrified collagen in multilayered microfluidic devices for corneal microtissue culture. Lab Chip 9, 3221–3227 (2009).

    Article  CAS  PubMed  Google Scholar 

  86. O'Neill, A.T., Monteiro-Riviere, N.A. & Walker, G.M. Characterization of microfluidic human epidermal keratinocyte culture. Cytotechnology 56, 197–207 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  87. Shin, M. et al. Endothelialized networks with a vascular geometry in microfabricated poly(dimethyl siloxane). Biomed. Microdevices 6, 269–278 (2004).

    Article  CAS  PubMed  Google Scholar 

  88. van der Meer, A.D., Orlova, V.V., ten Dijke, P., van den Berg, A. & Mummery, C.L. Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. Lab Chip 13, 3562–3568 (2013).

    Article  CAS  PubMed  Google Scholar 

  89. Shi, M. et al. Glia co-culture with neurons in microfluidic platforms promotes the formation and stabilization of synaptic contacts. Lab Chip 13, 3008–3021 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Park, H.S., Liu, S., McDonald, J., Thakor, N. & Yang, I.H. Neuromuscular junction in a microfluidic device. Conf. IEEE Eng. Med. Biol. Soc. 2013, 2833–2835 (2013).

    Google Scholar 

  91. Ziegler, L., Grigoryan, S., Yang, I.H., Thakor, N.V. & Goldstein, R.S. Efficient generation of schwann cells from human embryonic stem cell-derived neurospheres. Stem Cell Rev. 7, 394–403 (2011).

    Article  Google Scholar 

  92. Shayan, G., Choi, Y.S., Shusta, E.V., Shuler, M.L. & Lee, K.H. Murine in vitro model of the blood-brain barrier for evaluating drug transport. Eur. J. Pharm. Sci. 42, 148–155 (2011).

    Article  CAS  PubMed  Google Scholar 

  93. Shayan, G., Shuler, M.L. & Lee, K.H. The effect of astrocytes on the induction of barrier properties in aortic endothelial cells. Biotechnol. Prog. 27, 1137–1145 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. Achyuta, A.K. et al. A modular approach to create a neurovascular unit-on-a-chip. Lab Chip 13, 542–553 (2013).

    Article  CAS  PubMed  Google Scholar 

  95. Khetani, S.R. et al. The use of micropatterned co-cultures to detect compounds that cause drug induced liver injury in humans. Toxicol. Sci. 132, 107–117 (2013).

    Article  CAS  PubMed  Google Scholar 

  96. Khetani, S.R. & Bhatia, S.N. Microscale culture of human liver cells for drug development. Nat. Biotechnol. 26, 120–126 (2008).

    Article  CAS  PubMed  Google Scholar 

  97. Wood, D.K., et al. A biophysical indicator of vaso-occlusive risk in sickle cell disease. Sci. Transl. Med. 4, 123ra26 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Zhou, M., Ma, H., Lin, H. & Qin, J. Induction of epithelial-to-mesenchymal transition in proximal tubular epithelial cells on microfluidic devices. Biomaterials 35, 1390–1401 (2014).

    Article  CAS  PubMed  Google Scholar 

  99. Westein, E. et al. Atherosclerotic geometries exacerbate pathological thrombus formation poststenosis in a von Willebrand factordependent manner. Proc. Natl. Acad. Sci. USA 110, 1357–1362 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  101. Gopalan, S.M. et al. Anisotropic stretch-induced hypertrophy in neonatal ventricular myocytes micropatterned on deformable elastomers. Biotechnol. Bioeng. 81, 578–587 (2003).

    Article  CAS  PubMed  Google Scholar 

  102. McCain, M.L., Sheehy, S.P., Grosberg, A., Goss, J.A. & Parker, K.K. Recapitulating maladaptive, multiscale remodeling of failing myocardium on a chip. Proc. Natl. Acad. Sci. USA 110, 9770–9775 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. 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  PubMed  PubMed Central  Google Scholar 

  104. Li, C.Y. et al. Micropatterned cell-cell interactions enable functional encapsulation of primary hepatocytes in hydrogel microtissues. Tissue Eng. (in the press).

  105. Chen, A.A., Underhill, G.H. & Bhatia, S.N. Multiplexed, high-throughput analysis of 3D microtissue suspensions. Integr. Biol. (Camb) 2, 517–527 (2010).

    Article  CAS  Google Scholar 

  106. Chen, M.B., Whisler, J.A., Jeon, J.S. & Kamm, R.D. Mechanisms of tumor cell extravasation in an in vitro microvascular network platform. Integr. Biol. (Camb) 5, 1262–1271 (2013).

    Article  CAS  Google Scholar 

  107. Moya, M.L., Hsu, Y.H., Lee, A.P., Hughes, C.C. & George, S.C. In vitro perfused human capillary networks. Tissue Eng. Part C Methods 19, 730–737 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Bischel, L.L., Young, E.W., Mader, B.R. & Beebe, D.J. Tubeless microfluidic angiogenesis assay with three-dimensional endothelial-lined microvessels. Biomaterials 34, 1471–1477 (2013).

    Article  CAS  PubMed  Google Scholar 

  109. Hsu, Y.H., Moya, M.L., Hughes, C.C., George, S.C. & Lee, A.P. A microfluidic platform for generating large-scale nearly identical human microphysiological vascularized tissue arrays. Lab Chip 13, 2990–2998 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kunze, A. et al. Astrocyte-neuron co-culture on microchips based on the model of SOD mutation to mimic ALS. Integr. Biol. (Camb) 5, 964–975 (2013).

    Article  CAS  Google Scholar 

  111. Sung, K.E. et al. Transition to invasion in breast cancer: a microfluidic in vitro model enables examination of spatial and temporal effects. Integr. Biol. (Camb) 3, 439–450 (2011).

    Article  CAS  Google Scholar 

  112. Montanez-Sauri, S.I., Sung, K.E., Berthier, E. & Beebe, D.J. Enabling screening in 3D microenvironments: probing matrix and stromal effects on the morphology and proliferation of T47D breast carcinoma cells. Integr. Biol. (Camb) 5, 631–640 (2013).

    Article  CAS  Google Scholar 

  113. Lang, J.D., Berry, S.M., Powers, G.L., Beebe, D.J. & Alarid, E.T. Hormonally responsive breast cancer cells in a microfluidic co-culture model as a sensor of microenvironmental activity. Integr. Biol. (Camb) 5, 807–816 (2013).

    Article  CAS  Google Scholar 

  114. Song, J.W. et al. Microfluidic endothelium for studying the intravascular adhesion of metastatic breast cancer cells. PLoS ONE 4, e5756 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Wang, S. et al. Study on invadopodia formation for lung carcinoma invasion with a microfluidic 3D culture device. PLoS ONE 8, e56448 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zervantonakis, I.K. et al. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc. Natl. Acad. Sci. USA 109, 13515–13520 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Jeon, J.S., Zervantonakis, I.K., Chung, S., Kamm, R.D. & Charest, J.L. In vitro model of tumor cell extravasation. PLoS ONE 8, e56910 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Tatosian, D.A. & Shuler, M.L. A novel system for evaluation of drug mixtures for potential efficacy in treating multidrug resistant cancers. Biotechnol. Bioeng. 103, 187–198 (2009).

    Article  CAS  PubMed  Google Scholar 

  119. Sung, J.H. & Shuler, M.L. A micro cell culture analog (microCCA) with 3-D hydrogel culture of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer drugs. Lab Chip 9, 1385–1394 (2009).

    Article  CAS  PubMed  Google Scholar 

  120. Choucha Snouber, L. et al. Metabolomics-on-a-chip of hepatotoxicity induced by anticancer drug flutamide and Its active metabolite hydroxyflutamide using HepG2/C3a microfluidic biochips. Toxicol. Sci. 132, 8–20 (2013).

    Article  CAS  PubMed  Google Scholar 

  121. Shintu, L. et al. Metabolomics-on-a-chip and predictive systems toxicology in microfluidic bioartificial organs. Anal. Chem. 84, 1840–1848 (2012).

    Article  CAS  PubMed  Google Scholar 

  122. Choucha-Snouber, L. et al. Investigation of ifosfamide nephrotoxicity induced in a liver-kidney co-culture biochip. Biotechnol. Bioeng. 110, 597–608 (2013).

    Article  CAS  PubMed  Google Scholar 

  123. Gerets, H.H.J. et al. Characterization of primary human hepatocytes, HepG2 cells, and HepaRG cells at the mRNA level and CYP activity in response to inducers and their predictivity for the detection of human hepatotoxins. Cell Biol. Toxicol. 28, 69–87 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Wilkening, S., Stahl, F. & Bader, A. Comparison of primary human hepatocytes and hepatoma cell line Hepg2 with regard to their biotransformation properties. Drug Metab. Dispos. 31, 1035–1042 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Baudoin, R. et al. Evaluation of seven drug metabolisms and clearances by cryopreserved human primary hepatocytes cultivated in microfluidic biochips. Xenobiotica 43, 140–152 (2013).

    Article  CAS  PubMed  Google Scholar 

  126. Prot, J.M. et al. A cocktail of metabolic probes demonstrates the relevance of primary human hepatocyte cultures in a microfluidic biochip for pharmaceutical drug screening. Int. J. Pharm. 408, 67–75 (2011).

    Article  CAS  PubMed  Google Scholar 

  127. Chan, T. et al. Meeting the challenge of predicting hepatic clearance of compounds slowly metabolized by cytochrome P450 using a novel hepatocyte model, HepatoPac. Drug Metab. Dispos. 41, 2024–2032 (2013).

    Article  CAS  PubMed  Google Scholar 

  128. Baudoin, R. et al. Evaluation of a liver microfluidic biochip to predict in vivo clearances of seven drugs in rats. J. Pharm. Sci. 103, 706–718. (2014).

    Article  CAS  PubMed  Google Scholar 

  129. Thodeti, C.K. et al. TRPV4 channels mediate cyclic strain-induced endothelial cell reorientation through integrin-to-integrin signaling. Circ. Res. 104, 1123–1130 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Thorneloe, K.S. et al. An orally active TRPV4 channel blocker presents and resolves pulmonary edema induced by heart failure. Sci. Transl. Med. 4, 159ra148 (2012).

    Article  PubMed  CAS  Google Scholar 

  131. Faley, S.L. et al. Microfluidic single cell arrays to interrogate signalling dynamics of individual, patient-derived hematopoietic stem cells. Lab Chip 9, 2659–2664 (2009).

    Article  CAS  PubMed  Google Scholar 

  132. Xu, Z. et al. Application of a microfluidic chip-based 3D co-culture to test drug sensitivity for individualized treatment of lung cancer. Biomaterials 34, 4109–4117 (2013).

    Article  CAS  PubMed  Google Scholar 

  133. Allen, J.W., Khetani, S.R. & Bhatia, S.N. In vitro zonation and toxicity in a hepatocyte bioreactor. Toxicol. Sci. 84, 110–119 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  135. Kidambi, S. et al. Oxygen-mediated enhancement of primary hepatocyte metabolism, functional polarization, gene expression, and drug clearance. Proc. Natl. Acad. Sci. USA 106, 15714–15719 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  137. Lee, P.H. & Vandenburgh, H.H. Skeletal muscle atrophy in bioengineered skeletal muscle: a new model system. Tissue Eng. Part A 19, 2147–2155 (2013).

    Article  CAS  PubMed  Google Scholar 

  138. Rumsey, J.W., Das, M., Bhalkikar, A., Stancescu, M. & Hickman, J.J. Tissue engineering the mechanosensory circuit of the stretch reflex arc: sensory neuron innervation of intrafusal muscle fibers. Biomaterials 31, 8218–8227 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Ploss, A. et al. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc. Natl. Acad. Sci. USA 107, 3141–3145 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. March, S. et al. A microscale human liver platform that supports the hepatic stages of Plasmodium falciparum and vivax. Cell Host Microbe 14, 104–115 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Wagner, I. et al. A dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D human liver and skin tissue co-culture. Lab Chip 13, 3538–3547 (2013).

    Article  CAS  PubMed  Google Scholar 

  142. Domansky, K. et al. Perfused multiwell plate for 3D liver tissue engineering. Lab Chip 10, 51–58 (2010).

    Article  CAS  PubMed  Google Scholar 

  143. Sonntag, F. et al. Design and prototyping of a chip-based multi-micro-organoid culture system for substance testing, predictive to human (substance) exposure. J. Biotechnol. 148, 70–75 (2010).

    Article  CAS  PubMed  Google Scholar 

  144. Ataç, B. et al. Skin and hair on-a-chip: in vitro skin models versus ex vivo tissue maintenance with dynamic perfusion. Lab Chip 13, 3555–3561 (2013).

    Article  PubMed  CAS  Google Scholar 

  145. Schimek, K. et al. Integrating biological vasculature into a multi-organ-chip microsystem. Lab Chip 13, 3588–3598 (2013).

    Article  CAS  PubMed  Google Scholar 

  146. Günther, A. et al. A microfluidic platform for probing small artery structure and function. Lab Chip 10, 2341–2349 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Silva, P.N., Green, B.J., Altamentova, S.M. & Rocheleau, J.V. A microfluidic device designed to induce media flow throughout pancreatic islets while limiting shear-induced damage. Lab Chip 13, 4374–4384 (2013).

    Article  CAS  PubMed  Google Scholar 

  148. van Midwoud, P.M., Merema, M.T., Verpoorte, E. & Groothuis, G.M. A microfluidic approach for in vitro assessment of interorgan interactions in drug metabolism using intestinal and liver slices. Lab Chip 10, 2778–2786 (2010).

    Article  CAS  PubMed  Google Scholar 

  149. Scott, A. et al. A microfluidic microelectrode array for simultaneous electrophysiology, chemical stimulation, and imaging of brain slices. Lab Chip 13, 527–535 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Kaplowitz, N. Idiosyncratic drug hepatotoxicity. Nat. Rev. Drug Discov. 4, 489 (2005).

    Article  CAS  PubMed  Google Scholar 

  151. Domansky, K. et al. Clear castable polyurethane elastomer for fabrication of microfluidic devices. Lab Chip 13, 3956–3964 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Zhang, C. et al. Towards a human-on-chip: culturing multiple cell types on a chip with compartmentalized microenvironments. Lab Chip 9, 3185–3192 (2009).

    Article  CAS  PubMed  Google Scholar 

  153. Sung, J.H., Esch, M.B. & Shuler, M.L. Integration of in silico and in vitro platforms for pharmacokinetic-pharmacodynamic modeling. Expert Opin. Drug Metab. Toxicol. 6, 1063–1081 (2010).

    Article  CAS  PubMed  Google Scholar 

  154. Wikswo, J.P. et al. Scaling and systems biology for integrating multiple organsona-Chip. Lab Chip 13, 3496–3511 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Wikswo, J.P. et al. Engineering challenges for instrumenting and controlling integrated organ-on-chip systems. IEEE Trans. Biomed. Eng. 60, 682–690 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  156. Sung, J.H., Kam, C. & Shuler, M.L. A microfluidic device for a pharmacokinetic-pharmacodynamic (PK-PD) model on a chip. Lab Chip 10, 446–455 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Fleming, S. Khetani, D. Levner, G. Hamilton and T. Bahinski for their helpful input. This work was supported by grants from the Defense Advanced Research Projects Agency (DARPA) (W911NF-12-2-0036), the FDA (HHSF223201310079C), the US National Institutes of Health (UH3 EB017103, R01 DK85713, R01 EB008396), Bill and Melinda Gates Foundation (OPP1023607, OPP1086223) and the Wyss Institute for Biologically Inspired Engineering at Harvard University. S.N.B. is an investigator of the Howard Hughes Medical Institute; D.E.I. is a recipient of a US Department of Defense (DoD) Breast Cancer Innovator Award (BC074986).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Donald E Ingber.

Ethics declarations

Competing interests

S.N.B. is a co-founder of Hepregen Corporation, and D.E.I. is a founder of Emulate Inc.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bhatia, S., Ingber, D. Microfluidic organs-on-chips. Nat Biotechnol 32, 760–772 (2014). https://doi.org/10.1038/nbt.2989

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2989

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