Skip to main content
Top
Published in: Fluids and Barriers of the CNS 1/2019

Open Access 01-12-2019 | Research

An isogenic neurovascular unit model comprised of human induced pluripotent stem cell-derived brain microvascular endothelial cells, pericytes, astrocytes, and neurons

Authors: Scott G. Canfield, Matthew J. Stebbins, Madeline G. Faubion, Benjamin D. Gastfriend, Sean P. Palecek, Eric V. Shusta

Published in: Fluids and Barriers of the CNS | Issue 1/2019

Login to get access

Abstract

Background

Brain microvascular endothelial cells (BMECs) astrocytes, neurons, and pericytes form the neurovascular unit (NVU). Interactions with NVU cells endow BMECs with extremely tight barriers via the expression of tight junction proteins, a host of active efflux and nutrient transporters, and reduced transcellular transport. To recreate the BMEC-enhancing functions of NVU cells, we combined BMECs, astrocytes, neurons, and brain pericyte-like cells.

Methods

BMECs, neurons, astrocytes, and brain like pericytes were differentiated from human induced pluripotent stem cells (iPSCs) and placed in a Transwell-type NVU model. BMECs were placed in co-culture with neurons, astrocytes, and/or pericytes alone or in varying combinations and critical barrier properties were monitored.

Results

Co-culture with pericytes followed by a mixture of neurons and astrocytes (1:3) induced the greatest barrier tightening in BMECs, supported by a significant increase in junctional localization of occludin. BMECs also expressed active P-glycoprotein (PGP) efflux transporters under baseline BMEC monoculture conditions and continued to express baseline active PGP efflux transporters regardless of co-culture conditions. Finally, brain-like pericyte co-culture significantly reduced the rate of non-specific transcytosis across BMECs.

Conclusions

Importantly, each cell type in the NVU model was differentiated from the same donor iPSC source, yielding an isogenic model that could prove enabling for enhanced personalized modeling of the NVU in human health and disease.
Literature
2.
go back to reference Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37(1):13–25.PubMedCrossRef Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood–brain barrier. Neurobiol Dis. 2010;37(1):13–25.PubMedCrossRef
3.
go back to reference Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.PubMedCrossRef Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.PubMedCrossRef
4.
go back to reference Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16(1):1–13.PubMedCrossRef Ballabh P, Braun A, Nedergaard M. The blood–brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16(1):1–13.PubMedCrossRef
5.
go back to reference Daneman R, Agalliu D, Zhou L, Kuhnert F, Kuo CJ, Barres BA. Wnt/beta-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc Natl Acad Sci USA. 2009;106(2):641–6.PubMedCrossRefPubMedCentral Daneman R, Agalliu D, Zhou L, Kuhnert F, Kuo CJ, Barres BA. Wnt/beta-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc Natl Acad Sci USA. 2009;106(2):641–6.PubMedCrossRefPubMedCentral
6.
go back to reference Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature. 2010;468(7323):562–6.PubMedPubMedCentralCrossRef Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature. 2010;468(7323):562–6.PubMedPubMedCentralCrossRef
7.
go back to reference Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. The mouse blood–brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS ONE. 2010;5(10):e13741.PubMedPubMedCentralCrossRef Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. The mouse blood–brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS ONE. 2010;5(10):e13741.PubMedPubMedCentralCrossRef
8.
go back to reference Dohgu S, Takata F, Yamauchi A, et al. Brain pericytes contribute to the induction and up-regulation of blood–brain barrier functions through transforming growth factor-beta production. Brain Res. 2005;1038(2):208–15.PubMedCrossRef Dohgu S, Takata F, Yamauchi A, et al. Brain pericytes contribute to the induction and up-regulation of blood–brain barrier functions through transforming growth factor-beta production. Brain Res. 2005;1038(2):208–15.PubMedCrossRef
9.
go back to reference Weiss N, Miller F, Cazaubon S, Couraud PO. The blood–brain barrier in brain homeostasis and neurological diseases. Biochim Biophys Acta. 2009;1788(4):842–57.PubMedCrossRef Weiss N, Miller F, Cazaubon S, Couraud PO. The blood–brain barrier in brain homeostasis and neurological diseases. Biochim Biophys Acta. 2009;1788(4):842–57.PubMedCrossRef
10.
go back to reference Sweeney MD, Sagare AP, Zlokovic BV. blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14(3):133–50.PubMedPubMedCentralCrossRef Sweeney MD, Sagare AP, Zlokovic BV. blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol. 2018;14(3):133–50.PubMedPubMedCentralCrossRef
11.
go back to reference Vatine GD, Al-Ahmad A, Barriga BK, et al. Modeling psychomotor retardation using iPSCs from MCT8-deficient patients indicates a prominent role for the blood–brain barrier. Cell Stem Cell. 2017;20(6):831–43.PubMedPubMedCentralCrossRef Vatine GD, Al-Ahmad A, Barriga BK, et al. Modeling psychomotor retardation using iPSCs from MCT8-deficient patients indicates a prominent role for the blood–brain barrier. Cell Stem Cell. 2017;20(6):831–43.PubMedPubMedCentralCrossRef
12.
go back to reference Zlokovic BV. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201.PubMedCrossRef Zlokovic BV. The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178–201.PubMedCrossRef
13.
go back to reference Deli MA, Abrahám CS, Kataoka Y, Niwa M. Permeability studies on in vitro blood–brain barrier models: physiology, pathology, and pharmacology. Cell Mol Neurobiol. 2005;25(1):59–127.PubMedCrossRef Deli MA, Abrahám CS, Kataoka Y, Niwa M. Permeability studies on in vitro blood–brain barrier models: physiology, pathology, and pharmacology. Cell Mol Neurobiol. 2005;25(1):59–127.PubMedCrossRef
14.
go back to reference Syvänen S, Lindhe O, Palner M, et al. Species differences in blood–brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos. 2009;37(3):635–43.PubMedCrossRef Syvänen S, Lindhe O, Palner M, et al. Species differences in blood–brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos. 2009;37(3):635–43.PubMedCrossRef
15.
go back to reference Warren MS, Zerangue N, Woodford K, et al. Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human. Pharmacol Res. 2009;59(6):404–13.PubMedCrossRef Warren MS, Zerangue N, Woodford K, et al. Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human. Pharmacol Res. 2009;59(6):404–13.PubMedCrossRef
16.
go back to reference Weksler BB, Subileau EA, Perrière N, et al. blood–brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J. 2005;19(13):1872–4.PubMedCrossRef Weksler BB, Subileau EA, Perrière N, et al. blood–brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J. 2005;19(13):1872–4.PubMedCrossRef
17.
go back to reference Cecchelli R, Berezowski V, Lundquist S, et al. Modelling of the blood–brain barrier in drug discovery and development. Nat Rev Drug Discov. 2007;6(8):650–61.PubMedCrossRef Cecchelli R, Berezowski V, Lundquist S, et al. Modelling of the blood–brain barrier in drug discovery and development. Nat Rev Drug Discov. 2007;6(8):650–61.PubMedCrossRef
18.
go back to reference Calabria AR, Shusta EV. A genomic comparison of in vivo and in vitro brain microvascular endothelial cells. J Cereb Blood Flow Metab. 2008;28(1):135–48.PubMedCrossRef Calabria AR, Shusta EV. A genomic comparison of in vivo and in vitro brain microvascular endothelial cells. J Cereb Blood Flow Metab. 2008;28(1):135–48.PubMedCrossRef
19.
go back to reference Förster C, Burek M, Romero IA, Weksler B, Couraud PO, Drenckhahn D. Differential effects of hydrocortisone and TNFalpha on tight junction proteins in an in vitro model of the human blood–brain barrier. J Physiol. 2008;586(Pt 7):1937–49.PubMedPubMedCentralCrossRef Förster C, Burek M, Romero IA, Weksler B, Couraud PO, Drenckhahn D. Differential effects of hydrocortisone and TNFalpha on tight junction proteins in an in vitro model of the human blood–brain barrier. J Physiol. 2008;586(Pt 7):1937–49.PubMedPubMedCentralCrossRef
20.
go back to reference Man S, Ubogu EE, Williams KA, Tucky B, Callahan MK, Ransohoff RM. 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;2008:384982.PubMedPubMedCentralCrossRef Man S, Ubogu EE, Williams KA, Tucky B, Callahan MK, Ransohoff RM. 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;2008:384982.PubMedPubMedCentralCrossRef
21.
go back to reference Hollmann EK, Bailey AK, Potharazu AV, Neely MD, Bowman AB, Lippmann ES. Accelerated differentiation of human induced pluripotent stem cells to blood–brain barrier endothelial cells. Fluids Barriers CNS. 2017;14(1):9.PubMedPubMedCentralCrossRef Hollmann EK, Bailey AK, Potharazu AV, Neely MD, Bowman AB, Lippmann ES. Accelerated differentiation of human induced pluripotent stem cells to blood–brain barrier endothelial cells. Fluids Barriers CNS. 2017;14(1):9.PubMedPubMedCentralCrossRef
22.
go back to reference Lippmann ES, Azarin SM, Kay JE, et al. Derivation of blood–brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol. 2012;30(8):783–91.PubMedPubMedCentralCrossRef Lippmann ES, Azarin SM, Kay JE, et al. Derivation of blood–brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol. 2012;30(8):783–91.PubMedPubMedCentralCrossRef
23.
go back to reference Lippmann ES, Al-Ahmad A, Azarin SM, Palecek SP, Shusta EV. A retinoic acid-enhanced, multicellular human blood–brain barrier model derived from stem cell sources. Sci Rep. 2014;4:4160.PubMedPubMedCentralCrossRef Lippmann ES, Al-Ahmad A, Azarin SM, Palecek SP, Shusta EV. A retinoic acid-enhanced, multicellular human blood–brain barrier model derived from stem cell sources. Sci Rep. 2014;4:4160.PubMedPubMedCentralCrossRef
25.
go back to reference Canfield SG, Stebbins MJ, Morales BS, et al. An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells. J Neurochem. 2017;140(6):874–88.PubMedPubMedCentralCrossRef Canfield SG, Stebbins MJ, Morales BS, et al. An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells. J Neurochem. 2017;140(6):874–88.PubMedPubMedCentralCrossRef
26.
go back to reference Ribecco-Lutkiewicz M, Sodja C, Haukenfrers J, et al. A novel human induced pluripotent stem cell blood–brain barrier model: applicability to study antibody-triggered receptor-mediated transcytosis. Sci Rep. 2018;8(1):1873.PubMedPubMedCentralCrossRef Ribecco-Lutkiewicz M, Sodja C, Haukenfrers J, et al. A novel human induced pluripotent stem cell blood–brain barrier model: applicability to study antibody-triggered receptor-mediated transcytosis. Sci Rep. 2018;8(1):1873.PubMedPubMedCentralCrossRef
27.
go back to reference Lim RG, Quan C, Reyes-Ortiz AM, et al. Huntington’s disease iPSC-derived brain microvascular endothelial cells reveal WNT-mediated angiogenic and blood–brain barrier deficits. Cell Rep. 2017;19(7):1365–77.PubMedPubMedCentralCrossRef Lim RG, Quan C, Reyes-Ortiz AM, et al. Huntington’s disease iPSC-derived brain microvascular endothelial cells reveal WNT-mediated angiogenic and blood–brain barrier deficits. Cell Rep. 2017;19(7):1365–77.PubMedPubMedCentralCrossRef
28.
go back to reference DeStefano JG, Xu ZS, Williams AJ, Yimam N, Searson PC. Effect of shear stress on iPSC-derived human brain microvascular endothelial cells (dhBMECs). Fluids Barriers CNS. 2017;14(1):20.PubMedPubMedCentralCrossRef DeStefano JG, Xu ZS, Williams AJ, Yimam N, Searson PC. Effect of shear stress on iPSC-derived human brain microvascular endothelial cells (dhBMECs). Fluids Barriers CNS. 2017;14(1):20.PubMedPubMedCentralCrossRef
29.
go back to reference Katt ME, Linville RM, Mayo LN, Xu ZS, Searson PC. Functional brain-specific microvessels from iPSC-derived human brain microvascular endothelial cells: the role of matrix composition on monolayer formation. Fluids Barriers CNS. 2018;15(1):7.PubMedPubMedCentralCrossRef Katt ME, Linville RM, Mayo LN, Xu ZS, Searson PC. Functional brain-specific microvessels from iPSC-derived human brain microvascular endothelial cells: the role of matrix composition on monolayer formation. Fluids Barriers CNS. 2018;15(1):7.PubMedPubMedCentralCrossRef
30.
go back to reference Helms HC, Abbott NJ, Burek M, et al. 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. 2016;36(5):862–90.PubMedPubMedCentralCrossRef Helms HC, Abbott NJ, Burek M, et al. 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. 2016;36(5):862–90.PubMedPubMedCentralCrossRef
31.
go back to reference Brown JA, Pensabene V, Markov DA, et al. Recreating blood–brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics. 2015;9(5):054124.PubMedPubMedCentralCrossRef Brown JA, Pensabene V, Markov DA, et al. Recreating blood–brain barrier physiology and structure on chip: a novel neurovascular microfluidic bioreactor. Biomicrofluidics. 2015;9(5):054124.PubMedPubMedCentralCrossRef
32.
go back to reference Lippmann ES, Weidenfeller C, Svendsen CN, Shusta EV. Blood–brain barrier modeling with co-cultured neural progenitor cell-derived astrocytes and neurons. J Neurochem. 2011;119(3):507–20.PubMedPubMedCentralCrossRef Lippmann ES, Weidenfeller C, Svendsen CN, Shusta EV. Blood–brain barrier modeling with co-cultured neural progenitor cell-derived astrocytes and neurons. J Neurochem. 2011;119(3):507–20.PubMedPubMedCentralCrossRef
33.
go back to reference Qian T, Maguire SE, Canfield SG, et al. Directed differentiation of human pluripotent stem cells to blood–brain barrier endothelial cells. Sci Adv. 2017;3(11):e1701679.PubMedPubMedCentralCrossRef Qian T, Maguire SE, Canfield SG, et al. Directed differentiation of human pluripotent stem cells to blood–brain barrier endothelial cells. Sci Adv. 2017;3(11):e1701679.PubMedPubMedCentralCrossRef
34.
go back to reference Appelt-Menzel A, Cubukova A, Günther K, 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 Reports. 2017;8(4):894–906.PubMedPubMedCentralCrossRef Appelt-Menzel A, Cubukova A, Günther K, 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 Reports. 2017;8(4):894–906.PubMedPubMedCentralCrossRef
35.
go back to reference Xiang J, Andjelkovic AV, Wang MM, Keep RF. Blood–brain barrier models derived from individual patients: a new frontier: an Editorial Highlight on ‘An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells’. J Neurochem. 2017;140(6):843–4.PubMedCrossRef Xiang J, Andjelkovic AV, Wang MM, Keep RF. Blood–brain barrier models derived from individual patients: a new frontier: an Editorial Highlight on ‘An isogenic blood–brain barrier model comprising brain endothelial cells, astrocytes, and neurons derived from human induced pluripotent stem cells’. J Neurochem. 2017;140(6):843–4.PubMedCrossRef
36.
go back to reference Stebbins MJ, Gastfriend BD, Canfield SG, et al. Human pluripotent stem cell-derived brain pericyte-like cells induce blood–brain barrier properties. Sci Adv. 2019;5(3):eaau7375.PubMedPubMedCentralCrossRef Stebbins MJ, Gastfriend BD, Canfield SG, et al. Human pluripotent stem cell-derived brain pericyte-like cells induce blood–brain barrier properties. Sci Adv. 2019;5(3):eaau7375.PubMedPubMedCentralCrossRef
37.
go back to reference Ebert AD, Shelley BC, Hurley AM, et al. EZ spheres: a stable and expandable culture system for the generation of pre-rosette multipotent stem cells from human ESCs and iPSCs. Stem Cell Res. 2013;10(3):417–27.PubMedPubMedCentralCrossRef Ebert AD, Shelley BC, Hurley AM, et al. EZ spheres: a stable and expandable culture system for the generation of pre-rosette multipotent stem cells from human ESCs and iPSCs. Stem Cell Res. 2013;10(3):417–27.PubMedPubMedCentralCrossRef
38.
go back to reference Armulik A, Genové G, Mäe M, et al. Pericytes regulate the blood–brain barrier. Nature. 2010;468(7323):557–61.PubMedCrossRef Armulik A, Genové G, Mäe M, et al. Pericytes regulate the blood–brain barrier. Nature. 2010;468(7323):557–61.PubMedCrossRef
39.
go back to reference Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193–215.PubMedCrossRef Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193–215.PubMedCrossRef
40.
go back to reference Schiera G, Sala S, Gallo A, et al. Permeability properties of a three-cell type in vitro model of blood–brain barrier. J Cell Mol Med. 2005;9(2):373–9.PubMedPubMedCentralCrossRef Schiera G, Sala S, Gallo A, et al. Permeability properties of a three-cell type in vitro model of blood–brain barrier. J Cell Mol Med. 2005;9(2):373–9.PubMedPubMedCentralCrossRef
41.
go back to reference Nakagawa S, Deli MA, Kawaguchi H, et al. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54(3–4):253–63.PubMedCrossRef Nakagawa S, Deli MA, Kawaguchi H, et al. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54(3–4):253–63.PubMedCrossRef
42.
43.
go back to reference Wilson HK, Canfield SG, Hjortness MK, Palecek SP, Shusta EV. Exploring the effects of cell seeding density on the differentiation of human pluripotent stem cells to brain microvascular endothelial cells. Fluids Barriers CNS. 2015;12:13.PubMedPubMedCentralCrossRef Wilson HK, Canfield SG, Hjortness MK, Palecek SP, Shusta EV. Exploring the effects of cell seeding density on the differentiation of human pluripotent stem cells to brain microvascular endothelial cells. Fluids Barriers CNS. 2015;12:13.PubMedPubMedCentralCrossRef
44.
go back to reference Sareen D, Gowing G, Sahabian A, et al. Human induced pluripotent stem cells are a novel source of neural progenitor cells (iNPCs) that migrate and integrate in the rodent spinal cord. J Comp Neurol. 2014;522(12):2707–28.PubMedPubMedCentralCrossRef Sareen D, Gowing G, Sahabian A, et al. Human induced pluripotent stem cells are a novel source of neural progenitor cells (iNPCs) that migrate and integrate in the rodent spinal cord. J Comp Neurol. 2014;522(12):2707–28.PubMedPubMedCentralCrossRef
45.
go back to reference Azevedo FA, Carvalho LR, Grinberg LT, et al. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J Comp Neurol. 2009;513(5):532–41.PubMedCrossRef Azevedo FA, Carvalho LR, Grinberg LT, et al. Equal numbers of neuronal and nonneuronal cells make the human brain an isometrically scaled-up primate brain. J Comp Neurol. 2009;513(5):532–41.PubMedCrossRef
46.
go back to reference Herculano-Houzel S, Lent R. Isotropic fractionator: a simple, rapid method for the quantification of total cell and neuron numbers in the brain. J Neurosci. 2005;25(10):2518–21.PubMedPubMedCentralCrossRef Herculano-Houzel S, Lent R. Isotropic fractionator: a simple, rapid method for the quantification of total cell and neuron numbers in the brain. J Neurosci. 2005;25(10):2518–21.PubMedPubMedCentralCrossRef
47.
go back to reference van der Meer AD, Orlova VV, ten Dijke P, van den Berg A, Mummery CL. Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. Lab Chip. 2013;13(18):3562–8.PubMedCrossRef van der Meer AD, Orlova VV, ten Dijke P, van den Berg A, Mummery CL. Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. Lab Chip. 2013;13(18):3562–8.PubMedCrossRef
48.
go back to reference Kusuma S, Facklam A, Gerecht S. Characterizing human pluripotent-stem-cell-derived vascular cells for tissue engineering applications. Stem Cells Dev. 2015;24(4):451–8.PubMedCrossRef Kusuma S, Facklam A, Gerecht S. Characterizing human pluripotent-stem-cell-derived vascular cells for tissue engineering applications. Stem Cells Dev. 2015;24(4):451–8.PubMedCrossRef
49.
go back to reference Nordström U, Maier E, Jessell TM, Edlund T. An early role for WNT signaling in specifying neural patterns of Cdx and Hox gene expression and motor neuron subtype identity. PLoS Biol. 2006;4(8):e252.PubMedPubMedCentralCrossRef Nordström U, Maier E, Jessell TM, Edlund T. An early role for WNT signaling in specifying neural patterns of Cdx and Hox gene expression and motor neuron subtype identity. PLoS Biol. 2006;4(8):e252.PubMedPubMedCentralCrossRef
50.
go back to reference Maden M, Gale E, Kostetskii I, Zile M. Vitamin A-deficient quail embryos have half a hindbrain and other neural defects. Curr Biol. 1996;6(4):417–26.PubMedCrossRef Maden M, Gale E, Kostetskii I, Zile M. Vitamin A-deficient quail embryos have half a hindbrain and other neural defects. Curr Biol. 1996;6(4):417–26.PubMedCrossRef
51.
go back to reference Liu JP, Laufer E, Jessell TM. Assigning the positional identity of spinal motor neurons: rostrocaudal patterning of Hox-c expression by FGFs, Gdf11, and retinoids. Neuron. 2001;32(6):997–1012.PubMedCrossRef Liu JP, Laufer E, Jessell TM. Assigning the positional identity of spinal motor neurons: rostrocaudal patterning of Hox-c expression by FGFs, Gdf11, and retinoids. Neuron. 2001;32(6):997–1012.PubMedCrossRef
53.
go back to reference Stebbins MJ, Lippmann ES, Faubion MG, Daneman R, Palecek SP, Shusta EV. Activation of RARα, RARγ, or RXRα increases barrier tightness in human induced pluripotent stem cell-derived brain endothelial cells. Biotechnol J. 2018;13(2):1700093.CrossRef Stebbins MJ, Lippmann ES, Faubion MG, Daneman R, Palecek SP, Shusta EV. Activation of RARα, RARγ, or RXRα increases barrier tightness in human induced pluripotent stem cell-derived brain endothelial cells. Biotechnol J. 2018;13(2):1700093.CrossRef
54.
go back to reference Butt AM, Jones HC, Abbott NJ. Electrical resistance across the blood–brain barrier in anaesthetized rats: a developmental study. J Physiol. 1990;429:47–62.PubMedPubMedCentralCrossRef Butt AM, Jones HC, Abbott NJ. Electrical resistance across the blood–brain barrier in anaesthetized rats: a developmental study. J Physiol. 1990;429:47–62.PubMedPubMedCentralCrossRef
55.
go back to reference Weidenfeller C, Svendsen CN, Shusta EV. Differentiating embryonic neural progenitor cells induce blood–brain barrier properties. J Neurochem. 2007;101(2):555–65.PubMedPubMedCentralCrossRef Weidenfeller C, Svendsen CN, Shusta EV. Differentiating embryonic neural progenitor cells induce blood–brain barrier properties. J Neurochem. 2007;101(2):555–65.PubMedPubMedCentralCrossRef
56.
go back to reference Weidenfeller C, Schrot S, Zozulya A, Galla HJ. Murine brain capillary endothelial cells exhibit improved barrier properties under the influence of hydrocortisone. Brain Res. 2005;1053(1–2):162–74.PubMedCrossRef Weidenfeller C, Schrot S, Zozulya A, Galla HJ. Murine brain capillary endothelial cells exhibit improved barrier properties under the influence of hydrocortisone. Brain Res. 2005;1053(1–2):162–74.PubMedCrossRef
57.
go back to reference Calabria AR, Weidenfeller C, Jones AR, de Vries HE, Shusta EV. Puromycin-purified rat brain microvascular endothelial cell cultures exhibit improved barrier properties in response to glucocorticoid induction. J Neurochem. 2006;97(4):922–33.PubMedCrossRef Calabria AR, Weidenfeller C, Jones AR, de Vries HE, Shusta EV. Puromycin-purified rat brain microvascular endothelial cell cultures exhibit improved barrier properties in response to glucocorticoid induction. J Neurochem. 2006;97(4):922–33.PubMedCrossRef
58.
go back to reference Berezowski V, Landry C, Dehouck MP, Cecchelli R, Fenart L. Contribution of glial cells and pericytes to the mRNA profiles of P-glycoprotein and multidrug resistance-associated proteins in an in vitro model of the blood–brain barrier. Brain Res. 2004;1018(1):1–9.PubMedCrossRef Berezowski V, Landry C, Dehouck MP, Cecchelli R, Fenart L. Contribution of glial cells and pericytes to the mRNA profiles of P-glycoprotein and multidrug resistance-associated proteins in an in vitro model of the blood–brain barrier. Brain Res. 2004;1018(1):1–9.PubMedCrossRef
59.
go back to reference Perrière N, Yousif S, Cazaubon S, et al. A functional in vitro model of rat blood–brain barrier for molecular analysis of efflux transporters. Brain Res. 2007;1150:1–13.PubMedCrossRef Perrière N, Yousif S, Cazaubon S, et al. A functional in vitro model of rat blood–brain barrier for molecular analysis of efflux transporters. Brain Res. 2007;1150:1–13.PubMedCrossRef
60.
go back to reference Lim JC, Wolpaw AJ, Caldwell MA, Hladky SB, Barrand MA. Neural precursor cell influences on blood–brain barrier characteristics in rat brain endothelial cells. Brain Res. 2007;1159:67–76.PubMedCrossRef Lim JC, Wolpaw AJ, Caldwell MA, Hladky SB, Barrand MA. Neural precursor cell influences on blood–brain barrier characteristics in rat brain endothelial cells. Brain Res. 2007;1159:67–76.PubMedCrossRef
61.
go back to reference Freese C, Reinhardt S, Hefner G, Unger RE, Kirkpatrick CJ, Endres K. 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. 2014;9(3):e91003.PubMedPubMedCentralCrossRef Freese C, Reinhardt S, Hefner G, Unger RE, Kirkpatrick CJ, Endres K. 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. 2014;9(3):e91003.PubMedPubMedCentralCrossRef
62.
go back to reference Appelt-Menzel A, Cubukova A, Metzger M. Establishment of a human blood–brain barrier co-culture model mimicking the neurovascular unit using induced pluripotent stem cells. Curr Protoc Stem Cell Biol. 2018;47(1):e62.PubMedCrossRef Appelt-Menzel A, Cubukova A, Metzger M. Establishment of a human blood–brain barrier co-culture model mimicking the neurovascular unit using induced pluripotent stem cells. Curr Protoc Stem Cell Biol. 2018;47(1):e62.PubMedCrossRef
63.
go back to reference Alvarez JI, Dodelet-Devillers A, Kebir H, et al. The Hedgehog pathway promotes blood–brain barrier integrity and CNS immune quiescence. Science. 2011;334(6063):1727–31.PubMedCrossRef Alvarez JI, Dodelet-Devillers A, Kebir H, et al. The Hedgehog pathway promotes blood–brain barrier integrity and CNS immune quiescence. Science. 2011;334(6063):1727–31.PubMedCrossRef
64.
go back to reference Wosik K, Cayrol R, Dodelet-Devillers A, et al. Angiotensin II controls occludin function and is required for blood brain barrier maintenance: relevance to multiple sclerosis. J Neurosci. 2007;27(34):9032–42.PubMedPubMedCentralCrossRef Wosik K, Cayrol R, Dodelet-Devillers A, et al. Angiotensin II controls occludin function and is required for blood brain barrier maintenance: relevance to multiple sclerosis. J Neurosci. 2007;27(34):9032–42.PubMedPubMedCentralCrossRef
65.
go back to reference Nishitsuji K, Hosono T, Nakamura T, Bu G, Michikawa M. Apolipoprotein E regulates the integrity of tight junctions in an isoform-dependent manner in an in vitro blood–brain barrier model. J Biol Chem. 2011;286(20):17536–42.PubMedPubMedCentralCrossRef Nishitsuji K, Hosono T, Nakamura T, Bu G, Michikawa M. Apolipoprotein E regulates the integrity of tight junctions in an isoform-dependent manner in an in vitro blood–brain barrier model. J Biol Chem. 2011;286(20):17536–42.PubMedPubMedCentralCrossRef
66.
go back to reference Wang YI, Abaci HE, Shuler ML. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng. 2017;114(1):184–94.PubMedCrossRef Wang YI, Abaci HE, Shuler ML. Microfluidic blood–brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng. 2017;114(1):184–94.PubMedCrossRef
67.
Metadata
Title
An isogenic neurovascular unit model comprised of human induced pluripotent stem cell-derived brain microvascular endothelial cells, pericytes, astrocytes, and neurons
Authors
Scott G. Canfield
Matthew J. Stebbins
Madeline G. Faubion
Benjamin D. Gastfriend
Sean P. Palecek
Eric V. Shusta
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Fluids and Barriers of the CNS / Issue 1/2019
Electronic ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-019-0145-6

Other articles of this Issue 1/2019

Fluids and Barriers of the CNS 1/2019 Go to the issue