Skip to main content

Advertisement

Log in

A Reconfigurable In Vitro Model for Studying the Blood–Brain Barrier

  • Original Article
  • Published:
Annals of Biomedical Engineering Aims and scope Submit manuscript

Abstract

Much of what is currently known about the role of the blood–brain barrier (BBB) in regulating the passage of chemicals from the blood stream to the central nervous system (CNS) comes from animal in vivo models (requiring extrapolation to human relevance) and 2D static in vitro systems, which fail to capture the rich cell–cell and cell–matrix interactions of the dynamic 3D in vivo tissue microenvironment. In this work we have developed a BBB platform that allows for a high degree of customization in cellular composition, cellular orientation, and physiologically-relevant fluid dynamics. The system characterized and presented in this study reproduces key characteristics of a BBB model (e.g. tight junctions, efflux pumps) allowing for the formation of a selective and functional barrier. We demonstrate that our in vitro BBB is responsive to both biochemical and mechanical cues. This model further allows for culture of a CNS-like space around the BBB. The design of this platform is a valuable tool for studying BBB function as well as for screening of novel therapeutics.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Abbott, N. J., L. Rönnbäck, and E. Hansson. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 7:41, 2006.

    CAS  PubMed  Google Scholar 

  2. Adriani, G., D. Ma, A. Pavesi, R. D. Kamm, and E. L. K. Goh. A 3D neurovascular microfluidic model consisting of neurons, astrocytes and cerebral endothelial cells as a blood–brain barrier. Lab Chip 17:448–459, 2017.

    CAS  PubMed  Google Scholar 

  3. Appelt-Menzel, A., A. Cubukova, K. Günther, F. Edenhofer, J. Piontek, G. Krause, T. Stüber, H. Walles, W. Neuhaus, and M. Metzger. 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 

  4. Au-Yeung, K. L., K. Y. Sze, M. H. Sham, and B. P. Chan. Development of a micromanipulator-based loading device for mechanoregulation study of human mesenchymal stem cells in three-dimensional collagen constructs. Tissue Eng. C 16:93–107, 2010.

    CAS  Google Scholar 

  5. Bajaj, P., B. Reddy, Jr, L. Millet, C. Wei, P. Zorlutuna, G. Bao, and R. Bashir. Patterning the differentiation of C2C12 skeletal myoblasts. Integr. Biol. 3:897–909, 2011.

    CAS  Google Scholar 

  6. Bang, S., S.-R. Lee, J. Ko, K. Son, D. Tahk, J. Ahn, C. Im, and N. L. Jeon. 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 

  7. Banks, W. A. Developing drugs that can cross the blood–brain barrier: applications to Alzheimer’s disease. BMC Neurosci. 9:S2–S2, 2008.

    PubMed  PubMed Central  Google Scholar 

  8. Biemans, E. A. L. M., L. Jäkel, R. M. W. de Waal, H. B. Kuiperij, and M. M. Verbeek. Limitations of the hCMEC/D3 cell line as a model for Aβ clearance by the human blood–brain barrier. J. Neurosci. Res. 95:1513–1522, 2017.

    CAS  PubMed  Google Scholar 

  9. Birgersdotter, A., R. Sandberg, and I. Ernberg. Gene expression perturbation in vitro—a growing case for three-dimensional (3D) culture systems. Semin. Cancer Biol. 15:405–412, 2005.

    PubMed  Google Scholar 

  10. Booth, R., and H. Kim. Characterization of a microfluidic in vitro model of the blood–brain barrier (muBBB). Lab Chip 12:1784–1792, 2012.

    CAS  PubMed  Google Scholar 

  11. Brown, J. A., V. Pensabene, D. A. Markov, V. Allwardt, M. D. Neely, M. Shi, C. M. Britt, O. S. Hoilett, Q. Yang, B. M. Brewer, P. C. Samson, L. J. McCawley, J. M. May, D. J. Webb, D. Li, A. B. Bowman, R. S. Reiserer, and J. P. Wikswo. Recreating blood–brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics 9:054124, 2015.

    PubMed  PubMed Central  Google Scholar 

  12. Chistiakov, D. A., A. N. Orekhov, and Y. V. Bobryshev. Effects of shear stress on endothelial cells: go with the flow. Acta Physiol. 219:382–408, 2017.

    CAS  Google Scholar 

  13. Cho, C.-F., J. M. Wolfe, C. M. Fadzen, D. Calligaris, K. Hornburg, E. A. Chiocca, N. Y. R. Agar, B. L. Pentelute, and S. E. Lawler. Blood–brain-barrier spheroids as an in vitro screening platform for brain-penetrating agents. Nat. Commun. 8:15623, 2017.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Cucullo, L., N. Marchi, M. Hossain, and D. Janigro. A dynamic in vitro BBB model for the study of immune cell trafficking into the central nervous system. J. Cereb. Blood Flow Metab. 31:767–777, 2011.

    CAS  PubMed  Google Scholar 

  15. Deosarkar, S. P., B. Prabhakarpandian, B. Wang, J. B. Sheffield, B. Krynska, and M. F. Kiani. A novel dynamic neonatal blood–brain barrier on a chip. PLoS ONE 10:e0142725, 2015.

    PubMed  PubMed Central  Google Scholar 

  16. DeStefano, J. G., J. J. Jamieson, R. M. Linville, and P. C. Searson. Benchmarking in vitro tissue-engineered blood–brain barrier models. Fluids Barriers CNS 15:32, 2018.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. DeStefano, J. G., Z. S. Xu, A. J. Williams, N. Yimam, and P. C. Searson. Effect of shear stress on iPSC-derived human brain microvascular endothelial cells (dhBMECs). Fluids Barriers CNS 14:20, 2017.

    PubMed  PubMed Central  Google Scholar 

  18. Falanga, A. P., G. Pitingolo, M. Celentano, A. Cosentino, P. Melone, R. Vecchione, D. Guarnieri, and P. A. Netti. Shuttle-mediated nanoparticle transport across an in vitro brain endothelium under flow conditions. Biotechnol. Bioeng. 114:1087–1095, 2017.

    CAS  PubMed  Google Scholar 

  19. Förster, C., M. Burek, I. A. Romero, B. Weksler, P.-O. Couraud, and D. Drenckhahn. Differential effects of hydrocortisone and TNFα on tight junction proteins in an in vitro model of the human blood–brain barrier. J. Physiol. 586:1937–1949, 2008.

    PubMed  PubMed Central  Google Scholar 

  20. Garcia, P. A., J. H. Rossmeisl, J. L. Robertson, J. D. Olson, A. J. Johnson, T. L. Ellis, and R. V. Davalos. 70-T magnetic resonance imaging characterization of acute blood–brain-barrier disruption achieved with intracranial irreversible electroporation. PLoS ONE 7:e50482, 2012.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Griep, L. M., F. Wolbers, B. de Wagenaar, P. M. ter Braak, B. B. Weksler, I. A. Romero, P. O. Couraud, I. Vermes, A. D. van der Meer, and A. van den Berg. BBB ON CHIP: microfluidic platform to mechanically and biochemically modulate blood–brain barrier function. Biomed. Microdevices 15:145–150, 2013.

    CAS  PubMed  Google Scholar 

  22. Hatherell, K., P.-O. Couraud, I. A. Romero, B. Weksler, and G. J. Pilkington. 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 

  23. Helms, H. C., N. J. Abbott, M. Burek, R. Cecchelli, P.-O. Couraud, M. A. Deli, C. Förster, H. J. Galla, I. A. Romero, E. V. Shusta, M. J. Stebbins, E. Vandenhaute, B. Weksler, and B. Brodin. In vitro models of the blood–brain barrier: an overview of commonly used brain endothelial cell culture models and guidelines for their use. J. Cereb. Blood Flow Metab. 36:862–890, 2016.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Herland, A., A. D. van der Meer, E. A. FitzGerald, T.-E. Park, J. J. F. Sleeboom, and D. E. Ingber. 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 

  25. Hoosain, F. G., Y. E. Choonara, L. K. Tomar, P. Kumar, C. Tyagi, L. C. du Toit, and V. Pillay. Bypassing P-glycoprotein drug efflux mechanisms: possible applications in pharmacoresistant schizophrenia therapy. BioMed Res. Int. 2015:484963, 2015.

    PubMed  PubMed Central  Google Scholar 

  26. Hynynen, K., N. McDannold, N. A. Sheikov, F. A. Jolesz, and N. Vykhodtseva. Local and reversible blood–brain barrier disruption by noninvasive focused ultrasound at frequencies suitable for trans-skull sonications. NeuroImage 24:12–20, 2005.

    PubMed  Google Scholar 

  27. Jamieson, J. J., P. C. Searson, and S. Gerecht. Engineering the human blood–brain barrier in vitro. J. Biol. Eng. 11:37, 2017.

    PubMed  PubMed Central  Google Scholar 

  28. Jeong, S., S. Kim, J. Buonocore, J. Park, C. J. Welsh, J. Li, and A. Han. A three-dimensional arrayed microfluidic blood–brain barrier model with integrated electrical sensor array. IEEE Trans. Biomed. Eng. 65:431–439, 2018.

    PubMed  Google Scholar 

  29. Jiang, L., S. Li, J. Zheng, Y. Li, and H. Huang. Recent progress in microfluidic models of the blood–brain barrier. Micromachines (Basel) 2019. https://doi.org/10.3390/mi10060375.

    Article  Google Scholar 

  30. Kalvass, J. C., J. W. Polli, D. L. Bourdet, B. Feng, S.-M. Huang, X. Liu, Q. R. Smith, L. K. Zhang, and M. J. Zamek-Gliszczynski. Why clinical modulation of efflux transport at the human blood–brain barrier is unlikely: the ITC evidence-based position. Clin. Pharmacol. Ther. 94:80–94, 2013.

    CAS  PubMed  Google Scholar 

  31. Koo, Y., B. T. Hawkins, and Y. Yun. Three-dimensional (3D) tetra-culture brain on chip platform for organophosphate toxicity screening. Sci. Rep. 8:2841, 2018.

    PubMed  PubMed Central  Google Scholar 

  32. Liu, C., X.-N. Liu, G.-L. Wang, Y. Hei, S. Meng, L.-F. Yang, L. Yuan, and Y. Xie. A dual-mediated liposomal drug delivery system targeting the brain: rational construction, integrity evaluation across the blood–brain barrier, and the transporting mechanism to glioma cells. Int. J. Nanomed. 12:2407–2425, 2017.

    CAS  Google Scholar 

  33. Mairey, E., A. Genovesio, E. Donnadieu, C. Bernard, F. Jaubert, E. Pinard, J. Seylaz, J. C. Olivo-Marin, X. Nassif, and G. Dumenil. Cerebral microcirculation shear stress levels determine Neisseria meningitidis attachment sites along the blood–brain barrier. J. Exp. Med. 203:1939–1950, 2006.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Man, S., E. E. Ubogu, K. A. Williams, B. Tucky, M. K. Callahan, and R. M. Ransohoff. Human brain microvascular endothelial cells and umbilical vein endothelial cells differentially facilitate leukocyte recruitment and utilize chemokines for T cell migration. Clin. Dev. Immunol. 2008:384982, 2008.

    PubMed  PubMed Central  Google Scholar 

  35. Minagar, A., and J. S. Alexander. Blood–brain barrier disruption in multiple sclerosis. Mult. Scler. J. 9:540–549, 2003.

    CAS  Google Scholar 

  36. Nakagawa, S., M. A. Deli, H. Kawaguchi, T. Shimizudani, T. Shimono, Á. Kittel, K. Tanaka, and M. Niwa. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem. Int. 54:253–263, 2009.

    CAS  PubMed  Google Scholar 

  37. Nguyen, T. P. T., B. M. Tran, and N. Y. Lee. Microfluidic approach for the fabrication of cell-laden hollow fibers for endothelial barrier research. J. Mater. Chem. B 6:6057–6066, 2018.

    CAS  Google Scholar 

  38. Ni, Y., T. Teng, R. Li, A. Simonyi, G. Y. Sun, and J. C. Lee. TNFα alters occludin and cerebral endothelial permeability: role of p38MAPK. PLoS ONE 12:e0170346, 2017.

    PubMed  PubMed Central  Google Scholar 

  39. Odijk, M., A. D. Van Der Meer, D. Levner, H. J. Kim, M. W. Van Der Helm, L. I. Segerink, J. P. Frimat, G. A. Hamilton, D. E. Ingber, and A. Van Den Berg. Measuring direct current trans-epithelial electrical resistance in organ-on-a-chip microsystems. Lab Chip 15:745–752, 2015.

    CAS  PubMed  Google Scholar 

  40. Pardridge, W. M. The blood–brain barrier: bottleneck in brain drug development. NeuroRx 2:3–14, 2005.

    PubMed  PubMed Central  Google Scholar 

  41. Partyka, P. P., G. A. Godsey, J. R. Galie, M. C. Kosciuk, N. K. Acharya, R. G. Nagele, and P. A. Galie. Mechanical stress regulates transport in a compliant 3D model of the blood–brain barrier. Biomaterials 115:30–39, 2017.

    CAS  PubMed  Google Scholar 

  42. Persidsky, Y., S. H. Ramirez, J. Haorah, and G. D. Kanmogne. Blood–brain barrier: structural components and function under physiologic and pathologic conditions. J. Neuroimmune Pharmacol. 1:223–236, 2006.

    PubMed  Google Scholar 

  43. Prabhakarpandian, B., M.-C. Shen, J. B. Nichols, I. R. Mills, M. Sidoryk-Wegrzynowicz, M. Aschner, and K. Pant. SyM-BBB: a microfluidic blood brain barrier model. Lab Chip 13:1093–1101, 2013.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Reinitz, A., J. DeStefano, M. Ye, A. D. Wong, and P. C. Searson. Human brain microvascular endothelial cells resist elongation due to shear stress. Microvasc. Res. 99:8–18, 2015.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Saunders, N. R., M. D. Habgood, K. Møllgård, and K. M. Dziegielewska. The biological significance of brain barrier mechanisms: help or hindrance in drug delivery to the central nervous system? F1000Research 2016. https://doi.org/10.12688/f1000research.7378.1.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Sellgren, K. L., B. T. Hawkins, and S. Grego. An optically transparent membrane supports shear stress studies in a three-dimensional microfluidic neurovascular unit model. Biomicrofluidics 9:061102, 2015.

    PubMed  PubMed Central  Google Scholar 

  47. Shimokawa, H., and S. Godo. Diverse functions of endothelial NO synthases system: NO and EDH. J. Cardiovasc. Pharmacol. 67:361–366, 2016.

    CAS  PubMed  Google Scholar 

  48. Stanness, K. A., L. E. Westrum, E. Fornaciari, P. Mascagni, J. A. Nelson, S. G. Stenglein, T. Myers, and D. Janigro. Morphological and functional characterization of an in vitro blood–brain barrier model. Brain Res. 771:329–342, 1997.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Tibbe, M. P., A. M. Leferink, A. van den Berg, J. C. T. Eijkel, and L. I. Segerink. Microfluidic gel patterning method by use of a temporary membrane for organ-on-chip applications. Adv. Mater. Technol. 3:1700200, 2018.

    Google Scholar 

  51. Urich, E., C. Patsch, S. Aigner, M. Graf, R. Iacone, and P.-O. Freskgård. Multicellular self-assembled spheroidal model of the blood brain barrier. Sci. Rep. 3:1500, 2013.

    PubMed  PubMed Central  Google Scholar 

  52. van der Helm, M. W., A. D. van der Meer, J. C. T. Eijkel, A. van den Berg, and L. I. Segerink. Microfluidic organ-on-chip technology for blood–brain barrier research. Tissue Barriers 4:e1142493, 2016.

    PubMed  PubMed Central  Google Scholar 

  53. Walsby, E., A. Buggins, S. Devereux, C. Jones, G. Pratt, P. Brennan, C. Fegan, and C. Pepper. Development and characterization of a physiologically relevant model of lymphocyte migration in chronic lymphocytic leukemia. Blood 123:3607–3617, 2014.

    CAS  PubMed  Google Scholar 

  54. Wang, Y. I., H. E. Abaci, and M. L. Shuler. 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 

  55. Wang, J. D., E.-S. Khafagy, K. Khanafer, S. Takayama, and M. E. H. ElSayed. 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 

  56. Wei, X., X. Chen, M. Ying, and W. Lu. Brain tumor-targeted drug delivery strategies. Acta Pharm. Sin. B 4:193–201, 2014.

    PubMed  PubMed Central  Google Scholar 

  57. Weksler, B., I. A. Romero, and P.-O. Couraud. The hCMEC/D3 cell line as a model of the human blood brain barrier. Fluids Barriers CNS 10:16–16, 2013.

    PubMed  PubMed Central  Google Scholar 

  58. Ye, M., H. M. Sanchez, M. Hultz, Z. Yang, M. Bogorad, A. D. Wong, and P. C. Searson. Brain microvascular endothelial cells resist elongation due to curvature and shear stress. Sci. Rep. 4:4681, 2014.

    PubMed  PubMed Central  Google Scholar 

  59. Zhao, C., H. Wang, C. Xiong, and Y. Liu. Hypoxic glioblastoma release exosomal VEGF-A induce the permeability of blood–brain barrier. Biochem. Biophys. Res. Commun. 502:324–331, 2018.

    CAS  PubMed  Google Scholar 

  60. Zlokovic, B. V. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron 57:178–201, 2008.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was funded by LDRD Awards 14-SI-001 and 17-SI-002 under the auspices of the U.S. Department of Energy by Lawrence Livermore National Laboratory under Contract DE-AC52-07NA27344 Lawrence Livermore National Security, LLC. We acknowledge Scott Erickson, Sierra Levenson, Jonathan Adorno, and Haley Sandvik for their help with device fabrication efforts. LLNL-JRNL-758697.

Author information

Authors and Affiliations

Authors

Contributions

MM wrote the main manuscript text, analyzed the data and prepared all the figures. MM, RB, JA, MS, and MT assisted with design and fabrication of the devices. MM, JA and JO assisted with cell culture experiments. MT and DS designed the fluidic set-up. RB, FQ, MM provided input on initial device design. NF, KK and EW oversaw the project. All authors provided input for the manuscript.

Corresponding author

Correspondence to Monica L. Moya.

Ethics declarations

Conflict of interests

The authors declare no competing interests.

Additional information

Associate Editor Debra T. Auguste oversaw the review of this article.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moya, M.L., Triplett, M., Simon, M. et al. A Reconfigurable In Vitro Model for Studying the Blood–Brain Barrier. Ann Biomed Eng 48, 780–793 (2020). https://doi.org/10.1007/s10439-019-02405-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10439-019-02405-y

Keywords

Navigation