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.

  • Review Article
  • Published:

Human mini-brain models

Abstract

Engineered human mini-brains, made possible by knowledge from the convergence of precision microengineering and cell biology, permit systematic studies of complex neurological processes and of pathogenesis beyond what can be done with animal models. By culturing human brain cells with physiological microenvironmental cues, human mini-brain models reconstitute the arrangement of structural tissues and some of the complex biological functions of the human brain. In this Review, we highlight the most significant developments that have led to microphysiological human mini-brain models. We introduce the history of mini-brain development, review methods for creating mini-brain models in static conditions, and discuss relevant state-of-the-art dynamic cell-culture systems. We also review human mini-brain models that reconstruct aspects of major neurological disorders under static or dynamic conditions. Engineered human mini-brains will contribute to advancing the study of the physiology and aetiology of neurological disorders, and to the development of personalized medicines for them.

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: Neuronal damage mediated by dynamical mechanical cues leads to the alteration of neurovascular homeostasis.

Image courtesy of You Jung Kang, University of North Carolina at Charlotte

Fig. 2: Main techniques for the creation of human mini-brain models.
Fig. 3: Examples of mini-brain models using dynamic culture.
Fig. 4: Major neurological disorders recapitulated with mini-brain models under static or dynamic conditions.

Similar content being viewed by others

References

  1. Wyss-Coray, T. Ageing, neurodegeneration and brain rejuvenation. Nature 539, 180–186 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Abbott, N. J., Patabendige, A. A. K., Dolman, D. E. M., Yusof, S. R. & Begley, D. J. Structure and function of the blood–brain barrier. Neurobiol. Dis. 37, 13–25 (2010).

    CAS  PubMed  Google Scholar 

  3. Zlokovic, B. V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 12, 723–738 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Sturchler-Pierrat, C. et al. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc. Natl Acad. Sci. USA 94, 13287–13292 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  6. Ferdowsian, H. R. & Beck, N. Ethical and scientific considerations regarding animal testing and research. PLoS ONE 6, e24059 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

  8. Han, D. W. et al. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell 10, 465–472 (2012).

    CAS  PubMed  Google Scholar 

  9. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463, 1035–1041 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, P. et al. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol. Autism 8, 11 (2017).

    PubMed  PubMed Central  Google Scholar 

  11. Hardy, J. & Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci. 12, 383–388 (1991).

    CAS  PubMed  Google Scholar 

  12. Maries, E., Dass, B., Collier, T. J., Kordower, J. H. & Steece-Collier, K. The role of α-synuclein in Parkinson’s disease: insights from animal models. Nat. Rev. Neurosci. 4, 727–738 (2003).

    CAS  PubMed  Google Scholar 

  13. Visanji, N. P., Brooks, P. L., Hazrati, L. N. & Lang, A. E. The prion hypothesis in Parkinson’s disease: Braak to the future. Acta Neuropathol. Commun. 1, 2 (2013).

    PubMed  PubMed Central  Google Scholar 

  14. Brundin, P. & Melki, R. Prying into the prion hypothesis for Parkinson’s disease. J. Neurosci. 37, 9808–9818 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  16. Cucullo, L. et al. Immortalized human brain endothelial cells and flow-based vascular modeling: a marriage of convenience for rational neurovascular studies. J. Cereb. Blood Flow Metab. 28, 312–328 (2008).

    CAS  PubMed  Google Scholar 

  17. Booth, R., Noh, S. & Kim, H. A multiple-channel, multiple-assay platform for characterization of full-range shear stress effects on vascular endothelial cells. Lab Chip 14, 1880–1890 (2014).

    CAS  PubMed  Google Scholar 

  18. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, S., Wernig, M., Duncan, I. D. & Thomson, J. A. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nature 19, 1129–1133 (2001).

    CAS  Google Scholar 

  20. Campenot, R. B. Local control of neurite development by nerve growth factor. Proc. Natl Acad. Sci. USA 74, 4516–4519 (1977).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Rubin, L. L. et al. A cell culture model of the blood–brain barrier. J. Cell Biol. 115, 1725–1735 (1991).

    CAS  PubMed  Google Scholar 

  22. Patabendige, A., Skinner, R. A., Morgan, L. & Abbott, N. J. A detailed method for preparation of a functional and flexible blood–brain barrier model using porcine brain endothelial cells. Brain Res. 1521, 16–30 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Patabendige, A., Skinner, R. A. & Abbott, N. J. Establishment of a simplified in vitro porcine blood–brain barrier model with high transendothelial electrical resistance. Brain Res. 1521, 1–15 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Hatherell, K., Couraud, P. O., Romero, I. A., Weksler, B. & Pilkington, G. J. Development of a three-dimensional, all-human in vitro model of the blood–brain barrier using mono-, co-, and tri-cultivation Transwell models. J. Neurosci. Methods 199, 223–229 (2011).

    PubMed  Google Scholar 

  25. Freese, C. et al. A novel blood–brain barrier co-culture system for drug targeting of Alzheimer’s disease: establishment by using acitretin as a model drug. PLoS ONE 9, e91003 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Appelt-Menzel, A. et al. Establishment of a human blood–brain-barrier co-culture model mimicking the neurovascular unit using induced pluri- and multipotent stem cells. Stem Cell Rep. 8, 894–906 (2017).

    CAS  Google Scholar 

  27. Al-Shehri, A. et al. Permeability of PEGylated immunoarsonoliposomes through in vitro blood brain barrier-medulloblastoma co-culture models for brain tumor therapy. Pharm. Res. 32, 1072–1083 (2015).

    CAS  PubMed  Google Scholar 

  28. Biernacki, K., Prat, A., Blain, M. & Antel, J. P. Regulation of Th1 and Th2 lymphocyte migration by human adult brain endothelial cells. J. Neuropathol. Exp. Neurol. 60, 1127–1136 (2001).

    CAS  PubMed  Google Scholar 

  29. Thomsen, L. B., Burkhart, A. & Moos, T. A triple culture model of the blood–brain barrier using porcine brain endothelial cells, astrocytes and pericytes. PLoS ONE 10, e0134765 (2015).

    PubMed  PubMed Central  Google Scholar 

  30. Johnsen, K. B. et al. Targeting transferrin receptors at the blood–brain barrier improves the uptake of immunoliposomes and subsequent cargo transport into the brain parenchyma. Sci. Rep. 7, 10396 (2017).

    PubMed  PubMed Central  Google Scholar 

  31. Ruoslahti, E. Brain extracellular matrix. Glycobiology 6, 489–492 (1996).

    CAS  PubMed  Google Scholar 

  32. Dana, H. et al. Hybrid multiphoton volumetric functional imaging of large-scale bioengineered neuronal networks. Nat. Commun. 5, 3997 (2014).

    CAS  PubMed  Google Scholar 

  33. Thomson, D. Controlled growth en masse (somatic growth) of embryonic chick tissue in vitro. Proc. R. Soc. Med. 7, 71–75 (1914).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Aimetti, A. A., Machen, A. J. & Anseth, K. S. Poly(ethylene glycol) hydrogels formed by thiol-ene photopolymerization for enzyme-responsive protein delivery. Biomaterials 30, 6048–6054 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Birey, F. et al. Assembly of functionally integrated human forebrain spheroids. Nature 545, 54–59 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Reynolds, B. A., Tetzlaff, W. & Weiss, S. A multipotent EGF-responsive striatal embryonic progenitor cell produces neurons and astrocytes. J. Neurosci. 12, 4565–4574 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Eiraku, M. et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3, 519–532 (2008).

    CAS  PubMed  Google Scholar 

  38. Boutin, M. E. et al. A three-dimensional neural spheroid model for capillary-like network formation. J. Neurosci. Methods 299, 55–63 (2016).

    Google Scholar 

  39. Dingle, Y.-T. L. et al. Three-dimensional neural spheroid culture: an in vitro model for cortical studies. Tissue Eng. Part C Methods 21, 1274–1283 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Jo, J. et al. Midbrain-like organoids from human pluripotent stem cells contain functional dopaminergic and neuromelanin-producing neurons. Cell Stem Cell 19, 248–257 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  42. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    CAS  PubMed  Google Scholar 

  43. Quadrato, G. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48–53 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Camp, J. G. et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc. Natl Acad. Sci. USA 112, 15672–15677 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Qian, X. et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165, 1238–1254 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Velasco, S. et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 570, 523–527 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lancaster, M. A. et al. Guided self-organization and cortical plate formation in human brain organoids. Nat. Biotechnol. 35, 659–666 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Li, Y. et al. Induction of expansion and folding in human cerebral organoids. Cell Stem Cell 20, 385–396 (2017).

    PubMed  Google Scholar 

  49. Ogawa, J., Pao, G. M., Shokhirev, M. N. & Verma, I. M. Glioblastoma model using human cerebral organoids. Cell Rep. 23, 1220–1229 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Abbott, N. J. Blood–brain barrier structure and function and the challenges for CNS drug delivery. J. Inherit. Metab. Dis. 36, 437–449 (2013).

    CAS  PubMed  Google Scholar 

  51. Cui, H., Nowicki, M., Fisher, J. P. & Zhang, L. G. 3D Bioprinting for organ regeneration. Adv. Healthc. Mater. 6, 1601118 (2017).

    Google Scholar 

  52. Murphy, S. V. & Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 32, 773–785 (2014).

    CAS  PubMed  Google Scholar 

  53. Ravnic, D. J. et al. Transplantation of bioprinted tissues and organs technical and clinical challenges and future perspectives. Ann. Surg. 266, 48–58 (2017).

    PubMed  Google Scholar 

  54. Li, J., Chen, M., Fan, X. & Zhou, H. Recent advances in bioprinting techniques: approaches, applications and future prospects. J. Transl. Med. 14, 271 (2016).

    PubMed  PubMed Central  Google Scholar 

  55. Sanjana, N. E. & Fuller, S. B. A fast flexible ink-jet printing method for patterning dissociated neurons in culture. J. Neurosci. Methods 136, 151–163 (2004).

    PubMed  Google Scholar 

  56. Xu, T. et al. Viability and electrophysiology of neural cell structures generated by the inkjet printing method. Biomaterials 27, 3580–3588 (2006).

    CAS  PubMed  Google Scholar 

  57. Lee, Y. B. et al. Bio-printing of collagen and VEGF-releasing fibrin gel scaffolds for neural stem cell culture. Exp. Neurol. 223, 645–652 (2010).

    CAS  PubMed  Google Scholar 

  58. Lee, W. et al. Three-dimensional bioprinting of rat embryonic neural cells. NeuroReport 20, 798–803 (2009).

    PubMed  Google Scholar 

  59. Patz, T. M. et al. Three-dimensional direct writing of B35 neuronal cells. J. Biomed. Mater. Res. Part B Appl. Biomater. 78, 124–130 (2006).

    CAS  Google Scholar 

  60. Lozano, R. et al. 3D printing of layered brain-like structures using peptide modified gellan gum substrates. Biomaterials 67, 264–273 (2015).

    CAS  PubMed  Google Scholar 

  61. Hsieh, F. Y., Lin, H. H. & Hsu, S. 3D bioprinting of neural stem cell-laden thermoresponsive biodegradable polyurethane hydrogel and potential in central nervous system repair. Biomaterials 71, 48–57 (2015).

    CAS  PubMed  Google Scholar 

  62. Curley, J. L. et al. Isolated node engineering of neuronal systems using laser direct write. Biofabrication 8, 015013 (2016).

    CAS  PubMed  Google Scholar 

  63. Heinrich, M. A. et al. 3D-bioprinted mini-brain: a glioblastoma model to study cellular interactions and therapeutics. Adv. Mater. 31, e1806590 (2019).

    PubMed  Google Scholar 

  64. Gu, Q. et al. Functional 3D neural mini-tissues from printed gel-based bioink and human neural stem cells. Adv. Healthc. Mater. 5, 1429–1438 (2016).

    CAS  PubMed  Google Scholar 

  65. Abbott, N. J., Rönnbäck, L. & Hansson, E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 7, 41–53 (2006).

    CAS  PubMed  Google Scholar 

  66. Alrifaiy, A., Lindahl, O. A. & Ramser, K. Polymer-based microfluidic devices for pharmacy, biology and tissue engineering. Polymers (Basel) 4, 1349–1398 (2012).

    CAS  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  70. Chumbimuni-Torres, K. Y. et al. Adsorption of proteins to thin-films of PDMS and its effect on the adhesion of human endothelial cells. RSC Adv. 1, 706–714 (2011).

    CAS  PubMed  Google Scholar 

  71. Haring, A. P., Sontheimer, H. & Johnson, B. N. Microphysiological human brain and neural systems-on-a-chip: potential alternatives to small animal models and emerging platforms for drug discovery and personalized medicine. Stem Cell Rev. Rep. 13, 381–406 (2017).

    CAS  PubMed  Google Scholar 

  72. Taylor, A. M. et al. Microfluidic multicompartment device for neuroscience research. Langmuir 19, 1551–1556 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Taylor, A. M. et al. A microfluidic culture platform for CNS axonal injury, regeneration and transport. Nat. Methods 2, 599–605 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Yang, I. H., Siddique, R., Hosmane, S., Thakor, N. & Höke, A. Compartmentalized microfluidic culture platform to study mechanism of paclitaxel-induced axonal degeneration. Exp. Neurol. 218, 124–128 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Samson, A. J., Robertson, G., Zagnoni, M. & Connolly, C. N. Neuronal networks provide rapid neuroprotection against spreading toxicity. Sci. Rep. 6, 33746 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Taylor, A. M., Dieterich, D. C., Ito, H. T., Kim, S. A. & Schuman, E. M. Microfluidic local perfusion chambers for the visualization and manipulation of synapses. Neuron 66, 57–68 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Hengst, U., Deglincerti, A., Kim, H. J., Jeon, N. L. & Jaffrey, S. R. Axonal elongation triggered by stimulus-induced local translation of a polarity complex protein. Nat. Cell Biol. 11, 1024–1030 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Rivieccio, M. A. et al. HDAC6 is a target for protection and regeneration following injury in the nervous system. Proc. Natl Acad. Sci. USA 106, 19599–19604 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Kilic, O. et al. Brain-on-a-chip model enables analysis of human neuronal differentiation and chemotaxis. Lab Chip 16, 4152–4162 (2016).

    CAS  PubMed  Google Scholar 

  80. Wang, J. D., Khafagy, E. S., Khanafer, K., Takayama, S. & Elsayed, M. E. H. Organization of endothelial cells, pericytes, and astrocytes into a 3D microfluidic in vitro model of the blood–brain barrier. Mol. Pharm. 13, 895–906 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  82. 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).

    CAS  PubMed  Google Scholar 

  83. Cho, H. et al. Three-dimensional blood–brain barrier model for in vitro studies of neurovascular pathology. Sci. Rep. 5, 15222 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Xu, H. et al. A dynamic in vivo-like organotypic blood–brain barrier model to probe metastatic brain tumors. Sci. Rep. 6, 36670 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Adriani, G., Ma, D., Pavesi, A. & Kamm, R. D. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood–brain barrier. Lab Chip 12, 169–182 (2017).

    Google Scholar 

  86. Bang, S. et al. A low permeability microfluidic blood–brain barrier platform with direct contact between perfusable vascular network and astrocytes. Sci. Rep. 7, 8083 (2017).

    PubMed  PubMed Central  Google Scholar 

  87. Brown, J. A. et al. Recreating blood–brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics 9, 054124 (2015).

    PubMed  PubMed Central  Google Scholar 

  88. Herland, A. et al. Distinct contributions of astrocytes and pericytes to neuroinflammation identified in a 3D human blood–brain barrier on a chip. PLoS ONE 11, e0150360 (2016).

    PubMed  PubMed Central  Google Scholar 

  89. Park, T. E. et al. Hypoxia-enhanced Blood–Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat. Commun. 10, 2621 (2019).

    PubMed  PubMed Central  Google Scholar 

  90. Walter, F. R. et al. A versatile lab-on-a-chip tool for modeling biological barriers. Sens. Actuators B Chem. 222, 1209–1219 (2016).

    CAS  Google Scholar 

  91. Wang, Y. I., Abaci, H. E. & Shuler, M. L. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol. Bioeng. 114, 184–194 (2017).

    CAS  PubMed  Google Scholar 

  92. Seidel, D., Jahnke, H.-G., Englich, B., Girard, M. & Robitzki, A. A. In vitro field potential monitoring on a multi-microelectrode array for the electrophysiological long-term screening of neural stem cell maturation. Analyst 142, 1929–1937 (2017).

    CAS  PubMed  Google Scholar 

  93. Hofmann, F. & Bading, H. Long term recordings with microelectrode arrays: studies of transcription-dependent neuronal plasticity and axonal regeneration. J. Physiol. Paris 99, 125–132 (2006).

    CAS  PubMed  Google Scholar 

  94. Fan, Y., Nguyen, D. T., Akay, Y., Xu, F. & Akay, M. Engineering a brain cancer chip for high-throughput drug screening. Sci. Rep. 6, 25062 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Campisi, M. et al. 3D self-organized microvascular model of the human blood–brain barrier with endothelial cells, pericytes and astrocytes. Biomaterials 180, 117–129 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Park, J. et al. A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat. Neurosci. 21, 941–951 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Osaki, T., Sivathanu, V. & Kamm, R. D. Engineered 3D vascular and neuronal networks in a microfluidic platform. Sci. Rep. 8, 5168 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Lee, S. R. et al. Modeling neural circuit, blood–brain barrier, and myelination on a microfluidic 96 well plate. Biofabrication 11, 035013 (2019).

    CAS  PubMed  Google Scholar 

  99. Moreno, E. L. et al. Differentiation of neuroepithelial stem cells into functional dopaminergic neurons in 3D microfluidic cell culture. Lab Chip 15, 2419–2428 (2015).

    CAS  PubMed  Google Scholar 

  100. Wevers, N. R. et al. High-throughput compound evaluation on 3D networks of neurons and glia in a microfluidic platform. Sci. Rep. 6, 38856 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Shi, P. et al. Synapse microarray identification of small molecules that enhance synaptogenesis. Nat. Commun. 2, 510 (2011).

    PubMed  Google Scholar 

  102. Lee, J. N., Park, C. & Whitesides, G. M. Solvent compatibility of poly(dimethylsiloxane)-based microfluidic devices. Anal. Chem. 75, 6544–6554 (2003).

    CAS  PubMed  Google Scholar 

  103. Riaz, A. et al. Reactive deposition of nano-films in deep polymeric microcavities. Lab Chip 12, 4877 (2012).

    CAS  PubMed  Google Scholar 

  104. Abbott, N. J. Evidence for bulk flow of brain interstitial fluid: significance for physiology and pathology. Neurochem. Int. 45, 545–552 (2004).

    CAS  PubMed  Google Scholar 

  105. Park, J. et al. Three-dimensional brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer’s disease. Lab Chip 15, 141–150 (2014).

    Google Scholar 

  106. Choi, Y. J. et al. Neurotoxic amyloid beta oligomeric assemblies recreated in microfluidic platform with interstitial level of slow flow. Sci. Rep. 3, 1921 (2013).

    PubMed  PubMed Central  Google Scholar 

  107. Wong, A. D. et al. The blood–brain barrier: an engineering perspective. Front. Neuroeng. 6, 7 (2013).

    PubMed  PubMed Central  Google Scholar 

  108. Stanness, K. A. et al. Morphological and functional characterization of an in vitro blood–brain barrier model. Brain Res. 771, 329–342 (1997).

    CAS  PubMed  Google Scholar 

  109. Stanness, K. A., Guatteo, E. & Janigro, D. A dynamic model of the blood–brain barrier in vitro. Neurotoxicology 17, 481–496 (1996).

    CAS  PubMed  Google Scholar 

  110. Sei, Y., Justus, K., LeDuc, P. & Kim, Y. Engineering living systems on chips: from cells to human on chips. Microfluid. Nanofluid. 16, 907–920 (2014).

    CAS  Google Scholar 

  111. Vatine, G. D. et al. Human iPSC-derived blood–brain barrier chips enable disease modeling and personalized medicine applications. Cell Stem Cell 24, 995–1005 (2019).

    CAS  PubMed  Google Scholar 

  112. Wang, Y., Wang, L., Guo, Y., Zhu, Y. & Qin, J. Engineering stem cell-derived 3D brain organoids in a perfusable organ-on-a-chip system. RSC Adv. 8, 1677–1685 (2018).

    CAS  Google Scholar 

  113. Cakir, B. et al. Engineering of human brain organoids with a functional vascular-like system. Nat. Methods 16, 1169–1175 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Berdichevsky, Y., Staley, K. J. & Yarmush, M. L. Building and manipulating neural pathways with microfluidics. Lab Chip 10, 999–1004 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Hardy, J. & Selkoe, D. J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    CAS  PubMed  Google Scholar 

  117. Choi, S. H. et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274–278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Lee, H.-K. et al. Three dimensional human neuro-spheroid model of Alzheimer’s disease based on differentiated induced pluripotent stem cells. PLoS ONE 11, e0163072 (2016).

    PubMed  PubMed Central  Google Scholar 

  119. Raja, W. K. et al. Self-organizing 3D human neural tissue derived from induced pluripotent stem cells recapitulate Alzheimer’s disease phenotypes. PLoS ONE 11, e0161969 (2016).

    PubMed  PubMed Central  Google Scholar 

  120. Winklhofer, K. F. & Haass, C. Mitochondrial dysfunction in Parkinson’s disease. Biochim. Biophys. 1802, 29–44 (2010).

    CAS  Google Scholar 

  121. Song, H. L. et al. β-Amyloid is transmitted via neuronal connections along axonal membranes. Ann. Neurol. 75, 88–97 (2014).

    CAS  PubMed  Google Scholar 

  122. Deleglise, B. et al. β-Amyloid induces a dying-back process and remote trans-synaptic alterations in a microfluidic-based reconstructed neuronal network. Acta Neuropathol. Commun. 2, 145 (2014).

    PubMed  PubMed Central  Google Scholar 

  123. Poon, W. W. et al. β-Amyloid impairs axonal BDNF retrograde trafficking. Neurobiol. Aging 32, 821–833 (2011).

    CAS  PubMed  Google Scholar 

  124. Cho, H. et al. Microfluidic chemotaxis platform for differentiating the roles of soluble and bound amyloid-β on microglial accumulation. Sci. Rep. 3, 1823 (2013).

    PubMed  PubMed Central  Google Scholar 

  125. Dujardin, S. et al. Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies. Acta Neuropathol. Commun. 2, 14 (2014).

    PubMed  PubMed Central  Google Scholar 

  126. Calafate, S. et al. Synaptic contacts enhance cell-to-cell Tau pathology propagation. Cell Rep. 11, 1176–1183 (2015).

    CAS  PubMed  Google Scholar 

  127. Takeda, S. et al. Neuronal uptake and propagation of a rare phosphorylated high-molecular-weight tau derived from Alzheimer’s disease brain. Nat. Commun. 6, 8490 (2015).

    CAS  PubMed  Google Scholar 

  128. Wang, Y. et al. The release and trans-synaptic transmission of Tau via exosomes. Mol. Neurodegener. 12, 5 (2017).

    PubMed  PubMed Central  Google Scholar 

  129. Iliff, J. J. et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci. Transl. Med. 4, 147ra111 (2012).

    PubMed  PubMed Central  Google Scholar 

  130. Zhang, W. et al. Neuromelanin activates microglia and induces degeneration of dopaminergic neurons: implications for progression of Parkinson’s disease. Neurotox. Res. 19, 63–72 (2011).

    PubMed  Google Scholar 

  131. Zecca, L., Zucca, F. A., Wilms, H. & Sulzer, D. Neuromelanin of the substantia nigra: a neuronal black hole with protective and toxic characteristics. Trends Neurosci. 26, 578–580 (2003).

    CAS  PubMed  Google Scholar 

  132. Lee, H. J. et al. Dopamine promotes formation and secretion of non-fibrillar alpha-synuclein oligomers. Exp. Mol. Med. 43, 216–222 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Volpicelli-Daley, L. A. et al. Exogenous α-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 72, 57–71 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Hu, Q. & Wang, G. Mitochondrial dysfunction in Parkinson’s disease. Transl. Neurodegener. 5, 14 (2016).

    PubMed  PubMed Central  Google Scholar 

  135. Lu, X., Kim-Han, J. S., Malley, K. L. O. & Sakiyama-Elbert, S. E. A microdevice platform for visualizing mitochondrial transport in aligned dopaminergic axons. J. Neurosci. Methods 209, 35–39 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Huse, J. T. & Holland, E. C. Targeting brain cancer: advances in the molecular pathology of malignant glioma and medulloblastoma. Nat. Rev. Cancer 10, 319–331 (2010).

    CAS  PubMed  Google Scholar 

  137. Chonan, Y., Taki, S., Sampetrean, O., Saya, H. & Sudo, R. Endothelium-induced three-dimensional invasion of heterogeneous glioma initiating cells in a microfluidic coculture platform. Integr. Biol. 9, 762–773 (2017).

    CAS  Google Scholar 

  138. Beauchesne, P. Extra-neural metastases of malignant gliomas: myth or reality? Cancers (Basel) 3, 461–477 (2011).

    Google Scholar 

  139. Inglese, M. et al. Diffuse axonal injury in mild traumatic brain injury: a diffusion tensor imaging study. J. Neurosurg. 103, 298–303 (2005).

    PubMed  Google Scholar 

  140. Zetterberg, H., Smith, D. H. & Blennow, K. Biomarkers of mild traumatic brain injury in cerebrospinal fluid and blood. Nat. Rev. Neurol. 9, 201–210 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Sweeney, M. D., Sagare, A. P. & Zlokovic, B. V. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 14, 133–150 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Johnson, V. E. et al. Mechanical disruption of the blood–brain barrier following experimental concussion. Acta Neuropathol. 135, 711–726 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Bar-Kochba, E., Scimone, M. T., Estrada, J. B. & Franck, C. Strain and rate-dependent neuronal injury in a 3D in vitro compression model of traumatic brain injury. Sci. Rep. 6, 30550 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Tang-Schomer, M. D. et al. Bioengineered functional brain-like cortical tissue. Proc. Natl Acad. Sci. USA 111, 13811–13816 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Kanemaru, K. et al. Calcium-dependent N-cadherin up-regulation mediates reactive astrogliosis and neuroprotection after brain injury. Proc. Natl Acad. Sci. USA 110, 11612–11617 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Madathil, S. K. et al. Astrocyte-specific overexpression of insulin-like growth factor-1 protects hippocampal neurons and reduces behavioral deficits following traumatic brain injury in mice. PLoS ONE 8, e67204 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Moon, L. D. F. & Fawcett, J. W. Reduction in CNS scar formation without concomitant increase in axon regeneration following treatment of adult rat brain with a combination of antibodies to TGFβ1 and β2. Eur. J. Neurosci. 14, 1667–1677 (2001).

    CAS  PubMed  Google Scholar 

  148. Roth, T. L. et al. Transcranial amelioration of inflammation and cell death after brain injury. Nature 505, 223–228 (2014).

    CAS  PubMed  Google Scholar 

  149. Schachtrup, C. et al. Fibrinogen triggers astrocyte scar formation by promoting the availability of active TGF-β after vascular damage. J. Neurosci. 30, 5843–5854 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Gorina, R., Font-Nieves, M., Márquez-Kisinousky, L., Santalucia, T. & Planas, A. M. Astrocyte TLR4 activation induces a proinflammatory environment through the interplay between MyD88-dependent NFκB signaling, MAPK, and Jak1/Stat1 pathways. Glia 59, 242–255 (2011).

    PubMed  Google Scholar 

  151. Pedrazzi, M. et al. Selective proinflammatory activation of astrocytes by high mobility group Box 1 protein signaling. J. Immunol. 179, 8525–8532 (2007).

    CAS  PubMed  Google Scholar 

  152. Ponath, G. et al. Autocrine S100B effects on astrocytes are mediated via RAGE. J. Neuroimmunol. 184, 214–222 (2007).

    CAS  PubMed  Google Scholar 

  153. Maneshi, M. M., Sachs, F. & Hua, S. Z. A threshold shear force for calcium influx in an astrocyte model of traumatic brain injury. J. Neurotrauma 32, 1020–1029 (2015).

    PubMed  PubMed Central  Google Scholar 

  154. Mansour, A. A. et al. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 36, 432–441 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Ehsan, S. M., Welch-Reardon, K. M., Waterman, M. L., Hughes, C. C. W. & George, S. C. A three-dimensional in vitro model of tumor cell intravasation. Integr. Biol. 6, 603–610 (2014).

    CAS  Google Scholar 

  156. Wimmer, R. A. et al. Human blood vessel organoids as a model of diabetic vasculopathy. Nature 565, 505–510 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Kim, S. et al. Transneuronal propagation of pathologic α-synuclein from the gut to the brain models Parkinson’s disease. Neuron 103, 627–641 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Mass, E. et al. A somatic mutation in erythro-myeloid progenitors causes neurodegenerative disease. Nature 549, 389–393 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Zhang, W. et al. Aggregated α-synuclein activates microglia: a process leading to disease progression in Parkinson’s disease. FASEB J. 19, 533–542 (2005).

    CAS  PubMed  Google Scholar 

  160. Tang, T.-S. et al. Disturbed Ca2+ signaling and apoptosis of medium spiny neurons in Huntington’s disease. Proc. Natl Acad. Sci. USA 102, 2602–2607 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Presgraves, S. P., Ahmed, T., Borwege, S. & Joyce, J. N. Terminally differentiated SH-SY5Y cells provide a model system for studying neuroprotective effects of dopamine agonists. Neurotox. Res. 5, 579–598 (2004).

    PubMed  Google Scholar 

  162. Domert, J. et al. Neurobiology of disease spreading of amyloid-β peptides via neuritic cell-to-cell transfer is dependent on insufficient cellular clearance. Neurobiol. Dis. 65, 82–92 (2014).

    CAS  PubMed  Google Scholar 

  163. An, M. C. et al. Genetic correction of Huntington’s disease phenotypes in induced pluripotent stem cells. Cell Stem Cell 11, 253–263 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Dai, X., Ma, C., Lan, Q. & Xu, T. 3D bioprinted glioma stem cells for brain tumor model and applications of drug susceptibility. Biofabrication 8, 045005 (2016).

    PubMed  Google Scholar 

  165. Dai, X. et al. Coaxial 3D bioprinting of self-assembled multicellular heterogeneous tumor fibers. Sci. Rep. 7, 1457 (2017).

    PubMed  PubMed Central  Google Scholar 

  166. Terrell-Hall, T. B., Ammer, A. G., Griffith, J. I. G. & Lockman, P. R. Permeability across a novel microfluidic blood–tumor barrier model. Fluids Barriers CNS 14, 3 (2017).

    PubMed  PubMed Central  Google Scholar 

  167. Prabhakarpandian, B. et al. SyM-BBB: a microfluidic blood brain barrier model. Lab Chip 13, 1093–1101 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Maoz, B. M. et al. Organs-on-chips with combined multi-electrode array and transepithelial electrical resistance measurement capabilities. Lab Chip 17, 2294–2302 (2017).

    CAS  PubMed  Google Scholar 

  169. Osaki, T., Uzel, S. G. M. & Kamm, R. D. Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons. Sci. Adv. 4, eaat5847 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Shao, J. et al. Integrated microfluidic chip for endothelial cells culture and analysis exposed to a pulsatile and oscillatory shear stress. Lab Chip 9, 3118–3125 (2009).

    CAS  PubMed  Google Scholar 

  171. Yap, Y. C. et al. Mild and repetitive very mild axonal stretch injury triggers cystoskeletal mislocalization and growth cone collapse. PLoS ONE 12, e0176997 (2017).

    PubMed  PubMed Central  Google Scholar 

  172. Hosmane, S. et al. Toll/interleukin-1 receptor domain-containing adapter inducing interferon-β mediates microglial phagocytosis of degenerating axons. J. Neurosci. 32, 7745–7757 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Tang-Schomer, M. D., Davies, P., Graziano, D., Thurber, A. E. & Kaplan, D. L. Neural circuits with long-distance axon tracts for determining functional connectivity. J. Neurosci. Methods 222, 82–90 (2014).

    PubMed  Google Scholar 

  174. 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).

    PubMed  Google Scholar 

  175. Dollé, J.-P., Morrison, B. III, Schloss, R. S. & Yarmush, M. L. An organotypic uniaxial strain model using microfluidics. Lab Chip 13, 432–442 (2013).

    PubMed  Google Scholar 

  176. Tang, Y. T., Kim, J., Lopez-Valdes, H. E., Brennan, K. C. & Ju, Y. S. Microfluidic chamber with active suction ports for localized chemical stimulation of brain slices. Lab Chip 11, 2247–2254 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Karzbrun, E., Kshirsagar, A., Cohen, S. R., Hanna, J. H. & Reiner, O. Human brain organoids on a chip reveal the physics of folding. Nat. Phys. 14, 515–522 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Sei, Y. J., Ahn, S. I., Virtue, T., Kim, T. & Kim, Y. Detection of frequency-dependent endothelial response to oscillatory shear stress using a microfluidic transcellular monitor. Sci. Rep. 7, 10019 (2017).

    PubMed  PubMed Central  Google Scholar 

  179. Iadecola, C. & Davisson, R. L. Hypertension and cerebrovascular dysfunction. Cell Metab. 7, 476–484 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Lee, R. T. & Kamm, R. D. Vascular mechanics for the cardiologist. J. Am. Coll. Cardiol. 23, 1289–1295 (1994).

    CAS  PubMed  Google Scholar 

  181. Zhao, Z., Nelson, A. R., Betsholtz, C. & Zlokovic, B. V. Establishment and dysfunction of the blood–brain barrier. Cell 163, 1064–1078 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Cunningham, C., Dunne, A. & Lopez-Rodriguez, A. B. Astrocytes: heterogeneous and dynamic phenotypes in neurodegeneration and innate immunity. Neuroscientist 25, 455–474 (2018).

    PubMed  PubMed Central  Google Scholar 

  183. Hernandez-Ontiveros, D. G. et al. Microglia activation as a biomarker for traumatic brain injury. Front. Neurol. 4, 30 (2013).

    PubMed  PubMed Central  Google Scholar 

  184. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Napoli, I. & Neumann, H. Microglial clearance function in health and disease. Neuroscience 158, 1030–1038 (2009).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank W.-B. Tay (Ministry of Education, Singapore) and S. Neo (National University of Singapore) for proofreading the manuscript. This work was supported by the National Research Foundation (nos. NRF-2020R1A2C2010285 and NRF-2018M3C7A1056896 to H.C.)

Author information

Authors and Affiliations

Authors

Contributions

L.P.L. and H.C. shaped ideas and provided guidance. H.Y.T. and H.C. researched data for the article. H.Y.T. prepared the figures and wrote the manuscript with assistance from H.C. and L.P.L. All authors contributed to discussion of the contents, and reviewed and edited the manuscript.

Corresponding authors

Correspondence to Hansang Cho or Luke P. Lee.

Ethics declarations

Competing interests

The 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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tan, HY., Cho, H. & Lee, L.P. Human mini-brain models. Nat Biomed Eng 5, 11–25 (2021). https://doi.org/10.1038/s41551-020-00643-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-020-00643-3

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