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:

Astrocyte–endothelial interactions at the blood–brain barrier

Key Points

  • The blood–brain barrier (BBB), which is formed by the endothelial cells that line cerebral microvessels, has an important role in maintaining a precisely regulated microenvironment for reliable neuronal signalling. There is great interest in the association of brain microvessels, astrocytes and neurons to form functional 'neurovascular units', and recent studies have highlighted the importance of brain endothelial cells in this modular organization.

  • Three barrier layers limit and regulate molecular exchange at the interfaces between the blood and the neural tissue or its fluid spaces: the BBB between the blood and brain interstitial fluid, the choroid plexus epithelium between the blood and ventricular cerebrospinal fluid (CSF), and the arachnoid epithelium between the blood and subarachnoid CSF. Of these, the BBB exerts the greatest control over the immediate microenvironment of brain cells.

  • The BBB acts as a 'physical barrier' because complex tight junctions between adjacent endothelial cells force most molecular traffic to take a transcellular route. The presence of specific transport systems on the luminal and abluminal membranes regulates transcellular traffic, providing a selective 'transport barrier', and a combination of intracellular and extracellular enzymes allows the BBB to serve as a 'metabolic barrier'.

  • The BBB facilitates the entry of required nutrients into the brain, and excludes or effluxes potentially harmful compounds. It helps to keep separate the pools of neurotransmitters and neuroactive agents that act centrally and peripherally, and it regulates the ionic microenvironment of neurons.

  • Several features distinguish brain endothelium from the endothelium of most other tissues. In particular, the tight junctions are tighter and more complex in the brain endothelium. Among the molecules identified as making important contributions to tight junction structure are the transmembrane proteins occludin and the claudins. The brain endothelium also expresses several specific transport proteins at relatively high levels.

  • Brain capillaries are surrounded by or closely associated with several cell types, including neuronal processes and the perivascular endfeet of astrocytic glia, so it is not surprising to find synergistic inductive functions involving more than one cell type. For example, astrocytes secrete a range of factors that can induce aspects of the BBB phenotype in endothelial cells in vitro, and brain endothelium enhances the growth and differentiation of associated astrocytes.

  • Transmitters and modulators released by neurons, astrocytes and endothelium allow complex signalling between cells in the neurovascular unit, and many features of the BBB phenotype are subject to modulation under physiological or pathological conditions. For example, opening of the BBB's tight junctions may occur under normal conditions to allow the passage of growth factors and antibodies into the brain, and in inflammation can contribute to brain oedema.

  • Astrocytes occupy a strategic position between capillaries and neurons. Those that form perivascular endfeet at the BBB have a special role in ionic, amino acid, neurotransmitter and water homeostasis of the brain.

  • There is increasing evidence that the function of the BBB is altered in several neuropathologies, including brain oedema, epilepsy, Alzheimer's disease and Parkinson's disease. Damage to the endothelium could allow the expression of endothelial receptors that are normally downregulated, opening new communication loops between endothelium, pericytes, astrocytes and microglia that are important in barrier repair.

  • Reducing, halting or reversing BBB dysfunction could be of therapeutic value in conditions in which neuronal damage is secondary to or exacerbated by BBB damage. Moreover, maintaining endothelial health has the potential to delay or prevent the development of chronic neurodegeneration.

Abstract

The blood–brain barrier, which is formed by the endothelial cells that line cerebral microvessels, has an important role in maintaining a precisely regulated microenvironment for reliable neuronal signalling. At present, there is great interest in the association of brain microvessels, astrocytes and neurons to form functional 'neurovascular units', and recent studies have highlighted the importance of brain endothelial cells in this modular organization. Here, we explore specific interactions between the brain endothelium, astrocytes and neurons that may regulate blood–brain barrier function. An understanding of how these interactions are disturbed in pathological conditions could lead to the development of new protective and restorative therapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Location of barrier sites in the CNS.
Figure 2: Cellular constituents of the blood–brain barrier.
Figure 3: Pathways across the blood–brain barrier.
Figure 4: Molecular composition of endothelial tight junctions.
Figure 5: Complex cell–cell signalling at the blood–brain barrier.
Figure 6: Astroglial–endothelial signalling under pathological conditions.

Similar content being viewed by others

References

  1. Iadecola, C. Neurovascular regulation in the normal brain and in Alzheimer's disease. Nature Rev. Neurosci. 5, 347–360 (2004).

    Article  CAS  Google Scholar 

  2. Anderson, C. M. & Nedergaard, M. Astrocyte-mediated control of cerebral microcirculation. Trends Neurosci. 26, 340–344 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Nedergaard, M., Ransom, B. & Goldman, S. A. New roles for astrocytes: redefining the functional architecture of the brain. Trends Neurosci. 26, 523–530 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Davson, H. & Segal, M. B. Physiology of the CSF and Blood–Brain Barriers (CRC, Boca Raton, USA, 1995).

    Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  6. Schlageter, K. E., Molnar, P., Lapin, G. D. & Groothuis, D. R. Microvessel organization and structure in experimental brain tumors: microvessel populations with distinctive structural and functional properties. Microvasc. Res. 58, 312–328 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Risau, W. & Wolburg, H. Development of blood–brain barrier. Trends Neurosci. 13, 174–178 (1990).

    Article  CAS  PubMed  Google Scholar 

  8. Abbott, N. J. & Romero, I. A. Transporting therapeutics across the blood–brain barrier. Mol. Med. Today 2, 106–113 (1996).

    Article  CAS  PubMed  Google Scholar 

  9. Abbott, N. J. Astrocyte–endothelial interactions and blood–brain barrier permeability. J. Anat. 200, 629–638 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Begley, D. J. & Brightman, M. W. Structural and functional aspects of the blood–brain barrier. Prog. Drug Res. 61, 40–78 (2003).

    Google Scholar 

  11. Wolburg, H. & Lippoldt, A. Tight junctions of the blood–brain barrier: development, composition and regulation. Vasc. Pharmacol. 38, 323–337 (2002).

    Article  CAS  Google Scholar 

  12. Hawkins, B. T. & Davis, T. P. The blood–brain barrier/neurovascular unit in health and disease. Pharmacol. Rev. 57, 173–185 (2005).

    Article  CAS  PubMed  Google Scholar 

  13. El-Bacha, R. S. & Minn, A. Drug metabolizing enzymes in cerebrovascular endothelial cells afford a metabolic protection to the brain. Cell. Mol. Biol. 45, 15–23 (1999).

    CAS  PubMed  Google Scholar 

  14. Pardridge, W. M. Blood–brain barrier drug targeting: the future of brain drug development. Mol. Interv. 3, 90–105 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Ge, S., Song, L. & Pachter, J. S. Where is the blood–brain barrier...really? J. Neurosci. Res. 79, 421–427 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Abbott, N. J. Dynamics of CNS barriers: evolution, differentiation and modulation. Cell. Mol. Neurobiol. 25, 5–23 (2005).

    Article  PubMed  Google Scholar 

  17. Cserr, H. F. & Bundgaard, M. Blood–brain interfaces in vertebrates: a comparative approach. Am. J. Physiol. 246, R277–R288 (1984).

    Article  CAS  PubMed  Google Scholar 

  18. Brown, P. D., Davies, S. L., Speake, T. & Millar, I. D. Molecular mechanisms of cerebrospinal fluid production. Neuroscience 129, 957–970 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Chodobski, A. & Szmydynger-Chodobska, J. Choroid plexus: target for polypeptides and site of their synthesis. Microsc. Res. Tech. 52, 65–82 (2001).

    Article  CAS  PubMed  Google Scholar 

  20. Wolburg, H. in Blood–Brain Interfaces — from Ontogeny to Artificial Barriers (eds Dermietzel, R., Spray, D. & Nedergaard, M.) 77–107 (Wiley-VCH, Weinheim, Germany, in the press).

  21. Butt, A. M., Jones, H. C. & Abbott, N. J. Electrical resistance across the blood–brain barrier in anaesthetized rats: a developmental study. J. Physiol. (Lond.) 429, 47–62 (1990).

    Article  CAS  Google Scholar 

  22. Yu, A. et al. Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells. Am. J. Physiol. Cell Physiol. 288, 1231–1241 (2005).

    Article  CAS  Google Scholar 

  23. Simpson, I. A., Vanucci, S., DeJoseph, M. R. & Hawkins, R. A. Glucose transporter asymmetries in the bovine blood–brain barrier. J. Biol. Chem. 276, 12725–12729 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Schinkel, A. H. P-glycoprotein, a gatekeeper in the blood–brain barrier. Adv. Drug Deliv. Rev. 36, 179–194 (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Hawkins, R. A., Peterson, D. R. & Vina, J. R. The complementary membranes forming the blood–brain barrier. IUBMB Life 54, 101–107 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. O'Kane, R. & Hawkins, R. A. Na+-dependent transport of large neutral amino acids occurs at the abluminal membrane of the blood–brain barrier. Am. J. Physiol. Endocrinol. Metab. 285, E1167–E1173 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. O'Kane, R. L., Martinez-Lopez, I., DeJoseph, M. R., Vina, J. R. & Hawkins, R. A. Na+-dependent glutamate transporters (EAAT1, EAAT2, and EAAT3) of the blood–brain barrier. J. Biol. Chem. 274, 31891–31895 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Abbott, N. J. in Blood–Brain Interfaces — From Ontology to Artificial Barriers (eds Dermietzel, R., Spray, D. & Nedergaard, M.) 189–208 (Wiley-VCH, Weinheim, Germany, in the press).

  29. Davson, H. & Oldendorf, W. H. Transport in the central nervous system. Proc. R. Soc. Med. 60, 326–328 (1967).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Reichenbach, A. & Wolburg, H. in Neuroglia 2nd edn (eds Kettemann, H. & Ransom, B. R.) 19–35 (Oxford Univ. Press, New York, 2004).

    Book  Google Scholar 

  31. Dehouck, M. -P., Meresse, S., Delorme, P., Fruchart, J. C. & Cecchelli, R. An easier, reproducible, and mass-production method to study the blood–brain barrier in vitro. J. Neurochem. 54, 1798–1801 (1990). One of the first papers to describe a reliable method for generating an endothelial–astrocyte co-culture model of the BBB tight enough for study of permeability and transport. The model has since been successful in functional and mechanistic studies.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. McAllister, M. S. et al. Mechanisms of glucose transport at the blood–brain barrier: an in vitro study. Brain Res. 904, 20–30 (2001). Uses high-resolution confocal microscopy and permeability studies to show how perivascular astrocytes influence glucose transport by the brain endothelium.

    Article  CAS  PubMed  Google Scholar 

  34. Hayashi, Y. et al. Induction of various blood–brain barrier properties in non-neuronal endothelial cells by close apposition to co-cultured astrocytes. Glia 19, 13–26 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Sobue, K. et al. Induction of blood–brain barrier properties in immortalized bovine brain endothelial cells by astrocytic factors. Neurosci. Res. 35, 155–164 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Haseloff, R. F., Blasig, I. E., Bauer, H. -C. & Bauer, H. In search of the astrocytic factor(s) modulating blood–brain barrier functions in brain capillary endothelial cells in vitro. Cell. Mol. Neurobiol. 25, 25–39 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Duport, S. et al. An in vitro blood–brain barrier model: cocultures between endothelial cells and organotypic brain slice cultures. Proc. Natl Acad. Sci. USA 95, 1840–1845 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ramsauer, M., Krause, D. & Dermietzel, R. Angiogenesis of the blood–brain barrier in vitro and the function of cerebral pericytes. FASEB J. 16, 1274–1276 (2002). One of the first papers to study the complex interactions between endothelium, astrocytes and pericytes in vitro , giving insights into the development and maintenance of the neurovascular unit.

    Article  CAS  PubMed  Google Scholar 

  39. Zenker, D., Begley, D. J., Bratzke, H., Rübsamen-Waigmann, H. & von Briesen, H. Human blood-derived macrophages enhance barrier function of cultured primary bovine and human brain capillary endothelial cells. J. Physiol. (Lond.) 551, 1023–1032 (2003).

    Article  CAS  Google Scholar 

  40. Schiera, G. et al. Synergistic effects of neurons and astrocytes on the differentation of brain capillary endothelial cells in culture. J. Cell. Mol. Med. 7, 165–170 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Berezowski, V., Landry, C., Dehouck, M. P., 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. 1018, 1–9 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Hori, S., Ohtsuki, S., Hosoya, K., Nakashima, E. & Terasaki, T. A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro. J. Neurochem. 89, 503–513 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Dohgu, S. et al. Brain pericytes contribute to the induction and up-regulation of blood–brain barrier functions through transforming growth factor-β production. Brain Res. 1038, 208–215 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Estrada, C., Bready, J. V., Berliner, J. A., Pardridge, W. M. & Cancilla, P. A. Astrocyte growth stimulation by a soluble factor produced by cerebral endothelial cells in vitro. J. Neuropathol. Exp. Neurol. 49, 539–549 (1990).

    Article  CAS  PubMed  Google Scholar 

  45. Mi, H., Haeberle, H. & Barres, B. A. Induction of astrocyte differentiation by endothelial cells. J. Neurosci. 21, 1538–1547 (2001). An elegant study that used 'panning' to separate cell types from the optic nerve, showing convincingly that endothelium-derived LIF induces astrocyte differentiation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mizuguchi, H., Utoguchi, N. & Mayumi, T. Preparation of glial cell extracellular matrix: a novel method to analyze glial–endothelial interaction. Brain Res. Brain Res. Protoc. 1, 339–343 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Schroeter, M. L. et al. Astrocytes enhance radical defence in capillary endothelial cells constituting the blood–brain barrier. FEBS Lett. 449, 241–244 (1999). This co-culture study shows clearly the 'mutual induction' that astrocytes and endothelial cells exert on each other — free radical defence enzymes are upregulated in both cell types when they are grown together.

    Article  CAS  PubMed  Google Scholar 

  48. Verkman, A. S. Aquaporin water channels and endothelial cell function. J. Anat. 200, 617–627 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Garcia-Segura, L. M. & McCarthy, M. M. Minireview: role of glia in neuroendocrine function. Endocrinology 145, 1082–1086 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Igarashi, Y. et al. Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood–brain barrier. Biochem. Biophys. Res. Commun. 261, 108–112 (1999).

    Article  CAS  PubMed  Google Scholar 

  51. Lee, S. -W. et al. SSeCKS regulates angiogenesis and tight junction formation in blood–brain barrier. Nature Med. 9, 900–906 (2003). One of the most elegant and comprehensive studies of astrocyte–endothelial induction in vitro , revealing the novel role of Src-suppressor C-kinase substrate (SSeCKs) and angiopoetin 1.

    Article  CAS  PubMed  Google Scholar 

  52. Huber, J. D., Egleton, R. D. & Davis, T. P. Molecular physiology and pathophysiology of tight junctions in the blood–brain barrier. Trends Neurosci. 24, 719–725 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Drewes, L. R. in Introduction to the Blood–Brain Barrier — Methodology, Biology and Pathology (ed. Pardridge, W. M.) 165–174 (Cambridge Univ. Press, Cambridge, UK, 1998).

    Book  Google Scholar 

  54. Boado, R. J. & Pardridge, W. M. Glucose deprivation and hypoxia increase the expression of the GLUT-1 glucose transporter via a specific mRNA cis-acting regulatory element. J. Neurochem. 80, 552–554 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Pan, W., Akerstrom, V., Zhang, J., Pejovic, V. & Kastin, A. J. Modulation of feeding-related peptide/protein signals by the blood–brain barrier. J. Neurochem. 90, 455–461 (2004).

    Article  CAS  PubMed  Google Scholar 

  56. Abbott, N. J. Inflammatory mediators and modulation of blood–brain barrier permeability. Cell. Mol. Neurobiol. 20, 131–147 (2000).

    Article  CAS  PubMed  Google Scholar 

  57. Webb, A. A. & Muir, G. D. The blood–brain barrier and its role in inflammation. J. Vet. Intern. Med. 14, 399–411 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Tonra, J. R. Cerebellar susceptibility to experimental autoimmune encephalomyelitis in SJL/J mice: potential interaction of immunology with vascular anatomy. Cerebellum 1, 57–68 (2002).

    Article  PubMed  Google Scholar 

  59. Bauer, B., Hartz, A. M., Fricker, G. & Miller, D. S. Modulation of p-glycoprotein transport function at the blood–brain barrier. Exp. Biol. Med. 230, 118–127 (2005).

    Article  CAS  Google Scholar 

  60. Nwaozuzu, O. M., Sellers, L. A. & Barrand, M. A. Signalling pathways influencing basal and H2O2-induced P-glycoprotein expression in endothelial cells derived from the blood–brain barrier. J. Neurochem. 87, 1043–1051 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. Zhu, H. J. & Liu, G. Q. Glutamate up-regulates P-glycoprotein expression in rat brain microvessel endothelial cells by an NMDA receptor-mediated mechanism. Life Sci. 75, 1313–1322 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Bauer, B., Hartz, A. M. S., Fricker, G. & Miller, D. S. Pregnane X receptor up-regulation of P-glycoprotein expression and transport function at the blood–brain barrier. Mol. Pharmacol. 66, 413–419 (2004). Evidence that the nuclear PXR can upregulate Pgp function at the BBB, providing a mechanism for a number of long-term modulations of significance in physiology and pathology.

    CAS  PubMed  Google Scholar 

  63. Hartz, A. M. S., Bauer, B., Fricker, G. & Miller, D. S. Rapid regulation of P-glycoprotein at the blood–brain barrier by endothelin-1. Mol. Pharmacol. 66, 387–394 (2004). Complementary to reference 62, this paper shows short-term modulation of Pgp by a signalling molecule released within the neurovascular unit.

    Article  CAS  PubMed  Google Scholar 

  64. Hansson, E. & Rönnbäck, L. Astrocytic receptors and second messenger systems. Adv. Mol. Cell Biol. 31, 475–501 (2004).

    Article  CAS  Google Scholar 

  65. Hansson, E. & Rönnbäck, L. Astrocytes in glutamate neurotransmission. FASEB J. 9, 343–350 (1995).

    Article  CAS  PubMed  Google Scholar 

  66. Hansson, E. & Rönnbäck, L. Glial neuronal signalling in the central nervous system. FASEB J. 17, 341–348 (2003).

    Article  CAS  PubMed  Google Scholar 

  67. Andersson, A., Rönnbäck, L. & Hansson, E. Lactate induces tumour necrosis factor-α and interleukin-6 release in microglial and astroglial enriched primary cultures. J. Neurochem. 93, 1327–1333 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Kis, B. et al. Adrenomedullin regulates blood–brain barrier functions in vitro. Neuroreport 12, 4139–4142 (2001).

    Article  CAS  PubMed  Google Scholar 

  69. Brown, R. C., Mark, K. S., Egleton, R. D. & Davis, T. P. Protection against hypoxia-induced increase in blood–brain barrier permeability: role of tight junction proteins and NFκB. J. Cell Sci. 116, 693–700 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Mann, G. E., Yudilevich, D. L. & Sobrevia, L. Regulation of amino acid and glucose transport in endothelial and smooth muscle cells. Physiol. Rev. 83, 183–252 (2003).

    Article  CAS  PubMed  Google Scholar 

  71. Braet, K., Cabooter, L., Paemeleire, K. & Leybaert, L. Calcium signal communication in the central nervous system. Biol. Cell 96, 79–91 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Leybaert, L., Cabooter, L. & Braet, K. Calcium signal communication between glial and vascular brain cells. Acta Neurol. Belg. 104, 51–56 (2004).

    PubMed  Google Scholar 

  73. Leybaert, L. Neurobarrier coupling in the brain: a partner of neurovascular and neurometabolic coupling? J. Cereb. Blood Flow Metab. 25, 2–16 (2005).

    Article  CAS  PubMed  Google Scholar 

  74. Régina, A. et al. Factor(s) released by glucose-deprived astrocytes enhance glucose transporter expression and activity in rat brain endothelial cells. Biochim. Biophys. Acta 1540, 233–242 (2001). One of the first papers to show that the metabolic status of astrocytes affects the way they influence the brain endothelium, which is of relevance in ischaemia and starvation.

    Article  PubMed  Google Scholar 

  75. Abbott, N. J. in Introduction to the Blood–Brain Barrier: Methodology and Biology (ed. Pardridge, W. M.) 345–353 (Cambridge Univ. Press, Cambridge, UK, 1998).

    Book  Google Scholar 

  76. Muyderman, H. et al. α1-Adrenergic modulation of metabotropic glutamate receptor-induced calcium oscillations and glutamate release in astrocytes. J. Biol. Chem. 276, 46504–46514 (2001).

    Article  CAS  PubMed  Google Scholar 

  77. Pasti, L., Volterra, A., Pozzan, T. & Carmignoto, G. Intracellular calcium oscillations in astrocytes: a highly plastic, bidirectional form of communication between neurons and astrocytes in situ. J. Neurosci. 17, 7817–7830 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Cornell-Bell, A. H., Finkbeiner, S. M., Cooper, M. S. & Smith, S. J. Glutamate induces calcium waves in cultured astrocytes: long-range glial signalling. Science 247, 470–473 (1990).

    Article  CAS  PubMed  Google Scholar 

  79. Blomstrand, F. et al. 5-Hydroxytryptamine and glutamate modulate velocity and extent of intercellular calcium signalling in hippocampal astroglial cells in primary cultures. Neuroscience 88, 1241–1253 (1999).

    Article  CAS  PubMed  Google Scholar 

  80. Sneyd, J. et al. A model for the propagation of intercellular calcium waves. Am. J. Physiol. 266, C293–C302 (1994).

    Article  CAS  PubMed  Google Scholar 

  81. Cotrina, M. L. et al. Connexins regulate calcium signaling by controlling ATP release. Proc. Natl Acad. Sci. USA 95, 15735–15740 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Paemeleire, K. & Leybaert, L. ATP-dependent astrocyte–endothelial calcium signalling following mechanical damage to a single astrocyte in astrocyte–endothelial co-cultures. J. Neurotrauma 17, 345–358 (2000). One of the first papers to investigate the mechanisms that underlie rapid astrocyte–endothelial signalling, using cultured cells. It is now becoming possible to do this kind of experiment in brain slices.

    Article  CAS  PubMed  Google Scholar 

  83. Bezzi, P. et al. CXCR4-activated astrocyte glutamate release via TNFα: amplification by microglia triggers neurotoxicity. Nature Neurosci. 4, 702–710 (2001).

    Article  CAS  PubMed  Google Scholar 

  84. Rapoport, S. I. Blood–Brain Barrier in Physiology and Medicine (Raven, New York, USA, 1976).

    Google Scholar 

  85. Simard, M. & Nedergaard, M. The neurobiology of glia in the context of water and ion homeostasis. Neuroscience 129, 877–896 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Kofuji, P. & Newman, E. A. Potassium buffering in the central nervous system. Neuroscience 129, 1045–1056 (2004).

    Article  CAS  PubMed  Google Scholar 

  87. Price, D. L., Ludwig, J. W., Mi, H., Schwarz, T. L. & Ellisman, M. H. Distribution of rSlo Ca2+-activated K+ channels in rat astrocyte perivascular endfeet. Brain Res. 956, 183–193 (2002).

    Article  CAS  PubMed  Google Scholar 

  88. Simard, M., Arcuino, G., Takano, T., Liu, Q. S. & Nedergaard, M. Signaling at the gliovascular interface. J. Neurosci. 23, 9254–9262 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Amiry-Moghaddam, M. & Ottersen, O. P. The molecular basis of water transport in the brain. Nature Rev. Neurosci. 4, 991–1001 (2003).

    Article  CAS  Google Scholar 

  90. Dolman, D., Drndarski, S., Abbott, N. J. & Rattray, M. Induction of aquaporin 1 but not aquaporin 4 messenger RNA in rat primary brain microvessel endothelial cells in culture. J. Neurochem. 93, 825–833 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Hansson, E. Metabotropic glutamate receptor activation induces astroglial swelling. J. Biol. Chem. 269, 21955–21961 (1994).

    Article  CAS  PubMed  Google Scholar 

  92. Hansson, E., Johansson, B. B., Westergren, I. & Rönnbäck, L. Glutamate-induced swelling of single astroglial cells in primary culture. Neuroscience 63, 1057–1066 (1994).

    Article  CAS  PubMed  Google Scholar 

  93. Liebner, S. et al. Claudin-1 and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma multiforme. Acta Neuropathol. 100, 323–331 (2000).

    Article  CAS  PubMed  Google Scholar 

  94. Wolburg, H. et al. Localization of claudin-3 in tight junctions of the blood–brain barrier is selectively lost during experimental autoimmune encephalomyelitis and human glioblastoma multiforme. Acta Neuropathol. 105, 586–592 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Berzin, T. M. et al. Agrin and microvascular damage in Alzheimer's disease. Neurobiol. Aging 21, 349–355 (2000).

    Article  CAS  PubMed  Google Scholar 

  96. Warth, A., Kröger, S. & Wolburg, H. Redistribution of aquaporin-4 in human glioblastoma correlates with loss of agrin immunoreactivity from brain capillary basal laminae. Acta Neuropathol. 107, 311–318 (2004). Shows clearly the importance of the extracellular matrix in providing the scaffold for the ordering of proteins important in the function of astrocytic perivascular endfeet, and its disruption in pathology.

    Article  CAS  PubMed  Google Scholar 

  97. Minagar, A. & Alexander, J. S. Blood–brain barrier disruption in multiple sclerosis. Mult. Scler. 9, 540–549 (2003).

    Article  CAS  PubMed  Google Scholar 

  98. Abbott, N. J. et al. in Mechanisms of Drug Resistance in Epilepsy: Lessons from Oncology (ed. Ling, V.) Novartis Foundation Symposium No. 243, 38–47 (John Wiley, Chichester, UK, 2002).

    Google Scholar 

  99. Marroni, M. et al. Vascular and parenchymal mechanisms in multiple drug resistance: a lesson from human epilepsy. Curr. Drug Targets 4, 297–304 (2003).

    Article  CAS  PubMed  Google Scholar 

  100. Zlokovic, B. V. Neurovascular mechanisms of Alzheimer's neurodegeneration. Trends Neurosci. 28, 202–208 (2005).

    Article  CAS  PubMed  Google Scholar 

  101. Kortekaas, R. et al. Blood–brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol. 57, 176–179 (2005). An important but controversial paper showing how modern imaging techniques can be used to investigate BBB transport function in humans, and the insight this may give into disease states.

    Article  CAS  PubMed  Google Scholar 

  102. Schwaninger, M. et al. Bradykinin induces interleukin-6 expression in astrocytes through activation of nuclear factor-κB. J. Neurochem. 73, 1461–1466 (1999).

    Article  CAS  PubMed  Google Scholar 

  103. Deli, M. A. et al. Exposure of tumor necrosis factor-α to luminal membrane of bovine capillary endothelial cells cocultured with astrocytes induces a delayed increase of permeability and cytoplasmic stress formation of actine. J. Neurosci. Res. 41, 717–726 (1995).

    Article  CAS  PubMed  Google Scholar 

  104. Didier, N. et al. Secretion of interleukin-1β by astrocytes mediates endothelin-1 and tumour necrosis factor-α effects on human brain microvascular endothelial cell permeability. J. Neurochem. 86, 246–254 (2003). Illustrates the potential complexities of signalling between cells at the BBB — even apparently direct actions may involve indirect loops and potentiating (and inhibitory) modulation.

    Article  CAS  PubMed  Google Scholar 

  105. Perry, V. H., Newman, T. A. & Cunningham, C. The impact of systemic infection on the progression of neurodegenerative disease. Nature Rev. Neurosci. 4, 103–112 (2003).

    Article  CAS  Google Scholar 

  106. Banks, W. A. Blood–brain barrier transport of cytokines: a mechanism for neuropathology. Curr. Pharm. Des. 11, 973–984 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Watkins, L. R. & Maier, S. F. Glia: a novel drug discovery target for clinical pain. Nature Rev. Drug Discov. 2, 973–985 (2003).

    Article  CAS  Google Scholar 

  108. Huber, J. D. et al. Inflammatory pain alters blood–brain barrier permeability and tight junctional protein expression. Am. J. Physiol. Heart Circ. Physiol. 280, H1241–H1248 (2001). Recent work has shown, rather surprisingly, that even peripheral stimuli such as inflammatory pain can open the BBB.

    Article  CAS  PubMed  Google Scholar 

  109. Abbott, N. J. Prediction of blood–brain barrier permeation in drug discovery, from in vivo, in vitro and in silico models. Drug Discov. Today: Technologies 1, 407–416 (2004).

    Article  CAS  PubMed  Google Scholar 

  110. Dietrich, J. B. Endothelial cells of the blood–brain barrier: a target for glucocorticoids and estrogens? Front. Biosci. 9, 684–693 (2004).

    Article  CAS  PubMed  Google Scholar 

  111. Krizanac-Bengez, L., Mayberg, M. R. & Janigro, D. The cerebral vasculature as a therapeutic target for neurological disorders and the role of shear stress in vascular homeostasis and pathophysiology. Neurol. Res. 26, 846–853 (2004).

    Article  CAS  PubMed  Google Scholar 

  112. Demeule, M. et al. Brain endothelial cells as pharmacological targets in brain tumors. Mol. Neurobiol. 30, 157–183 (2004).

    Article  CAS  PubMed  Google Scholar 

  113. Kaal, E. C. & Vecht, C. J. The management of brain edema in brain tumors. Curr. Opin. Oncol. 16, 593–600 (2004).

    Article  CAS  PubMed  Google Scholar 

  114. Cucullo, L., Hallene, K., Dini, G., Dal Toso, R. & Janigro, D. Glycerophosphoinositol and dexamethasone improve transendothelial electrical resistance in an in vitro study of the blood–brain barrier. Brain Res. 997, 147–151 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Brown, R. C., Mark, K. S., Egleton, R. D. & Davis, T. P. Protection against hypoxia-induced blood–brain barrier disruption: changes in intracellular calcium. Am. J. Cell Physiol. 286, C1045–C1052 (2004).

    Article  CAS  Google Scholar 

  116. Turkel, N. A. & Ziylan, Z. Y. Protection of blood–brain barrier breakdown by nifedipine in adrenaline-induced hypertension. Int. J. Neurosci. 114, 517–528 (2004).

    Article  CAS  Google Scholar 

  117. Preston, E. & Webster, J. A two-hour window for hypothermic modulation of early events that impact delayed opening of the rat blood–brain barrier after ischemia. Acta Neuropathol. (Berl.) 108, 406–412 (2004).

    Article  Google Scholar 

  118. Wagner, K. R. & Zuccarello, M. Local brain hypothermia for neuroprotection in stroke treatment and aneurysm repair. Neurol. Res. 27, 238–245 (2005).

    Article  PubMed  Google Scholar 

  119. Park, S. et al. Neurovascular protection reduces early brain injury after subarachnoid hemorrhage. Stroke 35, 2412–2417 (2004).

    Article  CAS  PubMed  Google Scholar 

  120. Franzén, B. et al. Gene and protein expression profiling of human cerebral endothelial cells activated with tumor necrosis factor-α. Mol. Brain Res. 115, 130–146 (2003). With the human genome now fully sequenced, efforts are being made to identify genes and proteins of the brain endothelium that are activated in inflammation and disease, and that could therefore be useful targets for therapy and drug delivery to the brain. This is one of the first reports.

    Article  PubMed  CAS  Google Scholar 

  121. Kaya, D. et al. VEGF protects brain against focal ischemia without increasing blood–brain barrier permeability when administered intracerebro-ventricularly. J. Cereb. Blood Flow Metab. 25, 1111–1118 (2005).

    Article  CAS  PubMed  Google Scholar 

  122. Takahashi, M. & Macdonald, R. L. Vascular aspects of neuroprotection. Neurol. Res. 26, 862–869 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Rapoport, S. I. Advances in osmotic opening of the blood–brain barrier to enhance CNS chemotherapy. Expert Opin. Invest. Drugs 10, 1809–1818 (2001).

    Article  CAS  Google Scholar 

  124. Kraemer, D. F., Fortin, D. & Neuwelt, E. A. Chemotherapeutic dose intensification for treatment of malignant brain tumors: recent developments and future directions. Curr. Neurol. Neurosci. Rep. 2, 216–224 (2002).

    Article  PubMed  Google Scholar 

  125. Farkas, A. et al. Hyperosmotic mannitol induces Src kinase-dependent phosphorylation of β-catenin in cerebral endothelial cells. J. Neurosci. Res. 80, 855–861 (2005).

    Article  CAS  PubMed  Google Scholar 

  126. Prados, M. D. et al. A randomized, double-blind, placebo-controlled, phase 2 study of RMP-7 in combination with carboplatin administered intravenously for the treatment of recurrent malignant glioma. Neuro-oncology 5, 96–103 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ashraf, M. Z., Hussain, M. E. & Fahim, M. Antiatherosclerotic effects of dietary supplementations of garlic and turmeric: restoration of endothelial function in rats. Life. Sci. 77, 837–857 (2005).

    Article  CAS  Google Scholar 

  128. Rohdewald, P. A review of the French maritime pine bark extract (Pycnogenol), a herbal medication with a diverse clinical pharmacology. Int. J. Clin. Pharmacol. Ther. 40, 158–168 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Bijl, M. Endothelial activation, endothelial dysfunction, and premature atherosclerosis in systemic autoimmune diseases. Neth. J. Med. 61, 273–277 (2003).

    CAS  PubMed  Google Scholar 

  130. Calabresi, L., Gomaraschi, M. & Franceschini, G. Endothelial protection by high-density lipoproteins. From bench to bedside. Arterioscler. Thromb. Vasc. Biol. 23, 1724–1731 (2003).

    Article  CAS  PubMed  Google Scholar 

  131. d'Alessio, P. Aging and the endothelium. Exp. Gerontol. 39, 165–171 (2004).

    Article  CAS  PubMed  Google Scholar 

  132. Middlebrook, A. R. et al. Does aerobic fitness influence microvascular function in healthy adults at risk of developing type 2 diabetes? Diabet. Med. 22, 483–489 (2005).

    Article  Google Scholar 

  133. Abeywardena, M. Y. & Head, R. J. Longchain n-3 polyunsaturated fatty acids and blood vessel function. Cardiovasc. Res. 52, 361–371 (2001).

    Article  CAS  PubMed  Google Scholar 

  134. De Caterina, R., Madonna, R. & Massaro, M. Effects of omega-3 fatty acids on cytokines and adhesion molecules. Curr. Atheroscler. Rep. 6, 485–491 (2004).

    Article  PubMed  Google Scholar 

  135. Harris, H. W., Rockey, D. C., Young, D. M. & Welch, W. J. Diet-induced protection against lipopolysaccharide includes increased hepatic NO production. J. Surg. Res. 82, 339–345 (1999).

    Article  CAS  PubMed  Google Scholar 

  136. Kamata, K. et al. Effects of chronic administration of fruit extract (Citrus unshiu Marc) on endothelial dysfunction in streptozotocin-induced diabetic rats. Biol. Pharm. Bull. 28, 267–270 (2005).

    Article  CAS  PubMed  Google Scholar 

  137. Hwang, J., Hodis, H. N. & Sevanian, A. Soy and alfafa phytoestrogen extracts become potent low-density lipoprotein antioxidants in the presence of acerola cherry extract. J. Agric. Food Chem. 49, 308–314 (2001).

    Article  CAS  PubMed  Google Scholar 

  138. Vera, R. et al. Soy isoflavones improve endothelial function in spontaneously hypertensive rats in an estrogen-independent manner: role of nitric-oxide synthase, superoxide, and cyclooxygenase metabolites. J. Pharmacol. Exp. Ther. 314, 1300–1309 (2005).

    Article  CAS  PubMed  Google Scholar 

  139. d'Uscio, L. V., Milstein, S., Richardson, D., Smith, L. & Katusic, Z. S. Long-term vitamin C treatment increases vascular tetrahydrobiopterin levels and nitric oxide synthase activity. Circ. Res. 92, 88–95 (2003).

    Article  CAS  PubMed  Google Scholar 

  140. Marsh, S. A., Laursen, P. B., Pat, B. K., Gobe, G. C. & Coombes, J. J. Bcl-2 in endothelial cells is increased by vitamin E and α-lipoic acid supplementation but not exercise training. J. Mol. Cell. Cardiol. 38, 445–451 (2005).

    Article  CAS  PubMed  Google Scholar 

  141. Praticò, D. Antioxidants and endothelium protection. Atherosclerosis 181, 215–224 (2005).

    Article  PubMed  CAS  Google Scholar 

  142. Rasmussen, S. E., Frederiksen, H., Struntze Krogholm, K. & Poulsen, L. Dietary proanthocyanidins: occurrence, dietary intake, bioavailability, and protection aganist cardiovascular disease. Mol. Nutr. Food Res. 49, 159–174 (2005).

    Article  CAS  PubMed  Google Scholar 

  143. Fung, T. T. et al. Diet-quality scores and plasma concentrations of markers of inflammation and endothelial dysfunction. Am. J. Clin. Nutr. 82, 163–173 (2005). This study on the effect of diet on systemic endothelial function is a useful indicator of possible ways to maintain a healthy BBB.

    Article  CAS  PubMed  Google Scholar 

  144. Youdim, K. A., Spencer, J. P., Schroeter, H. & Rice-Evans, C. Dietary flavonoids as potential neuroprotectants. Biol. Chem. 383, 503–519 (2002).

    Article  CAS  PubMed  Google Scholar 

  145. Yoshida, H. et al. Inhibitory effect of tea flavonoids on the ability of cells to oxidize low density lipoprotein. Biochem. Pharmacol. 58, 1695–1703 (1999).

    Article  CAS  PubMed  Google Scholar 

  146. Stoclet, J. C. et al. Vascular protection by dietary polyphenols. Eur. J. Pharmacol. 500, 299–313 (2004).

    Article  CAS  PubMed  Google Scholar 

  147. Kawakami, M., Sekiguchi, M., Sato, K., Kozaki, S. & Takahashi, M. Erythropoietin receptor-mediated inhibition of exocytotic glutamate release confers neuroprotection during chemical ischemia. J. Biol. Chem. 276, 39469–39475 (2001).

    Article  CAS  PubMed  Google Scholar 

  148. Martínez-Estrada, O. M. et al. Erythropoietin protects the in vitro blood–brain barrier against VEGF-induced permeability. Eur. J. Neurosci. 18, 2538–2544 (2003).

    Article  PubMed  Google Scholar 

  149. Zonta, M. et al. Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation. Nature Neurosci. 6, 43–50 (2003).

    Article  CAS  PubMed  Google Scholar 

  150. Tran, N. D., Correale, J., Schrieber, S. S. & Fisher, M. Transforming growth factor-β mediates astrocyte-specific regulation of brain endothelial anticoagulant factors. Stroke 30, 1671–1677 (1999).

    Article  CAS  PubMed  Google Scholar 

  151. Lo, E. H., Dalkara, T. & Moskowitz, M. A. Mechanisms, challenges and opportunities in stroke. Nature Rev. Neurosci. 4, 399–415 (2003).

    Article  CAS  Google Scholar 

  152. Tomas-Camardiel, M. et al. Blood–brain barrier disruption highly induces aquaporin-4 mRNA and protein in perivascular and parenchymal astrocytes: protective effect by estradiol treatment in ovariectomized animals. J. Neurosci. Res. 80, 235–246 (2005).

    Article  CAS  PubMed  Google Scholar 

  153. Vakili, A., Kataoka, H. & Plesnila, N. Role of arginine vasopressin V1 and V2 receptors for brain damage after transient focal cerebral ischemia. J. Cereb. Blood Flow Metab. 25, 1012–1019 (2005).

    Article  CAS  PubMed  Google Scholar 

  154. Gaillard, P. J., de Boer, A. B. & Breimer, D. D. Pharmacological investigations on lipopolysaccharide-induced permeability changes in the blood–brain barrier in vitro. Microvasc. Res. 65, 24–31 (2003).

    Article  CAS  PubMed  Google Scholar 

  155. Veldhuis, W. B. et al. Interferon-β prevents cytokine-induced neutrophil infiltration and attenuates blood–brain barrier disruption. J. Cereb. Blood Flow Metab. 23, 1060–1069 (2003).

    Article  CAS  PubMed  Google Scholar 

  156. Oki, T. et al. Increased ability of peripheral blood lymphocytes to degrade laminin in multiple sclerosis. J. Neurol. Sci. 222, 7–11 (2004).

    Article  CAS  PubMed  Google Scholar 

  157. Dallasta, L. M. et al. Blood–brain barrier tight junction disruption in human immunodeficiency virus-1 encephalitis. Am. J. Pathol. 155, 1915–1927 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Berger, J. R. & Avison, M. The blood brain barrier in HIV infection. Front. Biosci. 9, 2680–2685 (2004).

    Article  CAS  PubMed  Google Scholar 

  159. Kalaria, R. N. The blood–brain barrier and cerebrovascular pathology in Alzheimer's disease. Ann. NY Acad. Sci. 893, 113–125 (1999).

    Article  CAS  PubMed  Google Scholar 

  160. Lee, G. & Bendayan, R. Functional expression and localization of P-glycoprotein in the central nervous system: relevance to the pathogenesis and treatment of neurological disorders. Pharm. Res. 21, 1313–1320 (2004).

    Article  CAS  PubMed  Google Scholar 

  161. Papadopoulos, M. C., Saadoun, S., Davies, D. C. & Bell, B. A. Emerging molecular mechanisms of brain tumour oedema. Br. J. Neurosurg. 15, 101–108 (2001).

    Article  CAS  PubMed  Google Scholar 

  162. Davies, D. C. Blood–brain barrier breakdown in septic encephalopathy and brain tumours. J. Anat. 200, 639–646 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Segal, M. B. & Zlokovic, B. V. The Blood–Brain Barrier, Amino Acids and Peptides (Kluwer Academic, Dordrecht, Boston (USA) & London (UK), 1990).

    Google Scholar 

Download references

Acknowledgements

The work performed in the authors' laboratories was supported by grants from the Wellcome Trust (N.J.A.) and the Swedish Research Council (L.R. and E.H.). Figures 2 and 6 were originally designed by E. Kraft, Göteborg, Sweden, who is gratefully acknowledged.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to N. Joan Abbott.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Abbott's homepage

Glossary

Neurovascular unit

A functional unit composed of groups of neurons and their associated astrocytes, interacting with smooth muscle cells and endothelial cells on the microvessels (arterioles) responsible for their blood supply, and capable of regulating the local blood flow.

Gliovascular unit

A proposed functional unit composed of single astrocytic glial cells and the neurons they surround, interacting with local segments of blood vessels, and capable of regulating blood flow at the arteriolar level and BBB functions at the capillary level.

Choroid plexus

A site of production of CSF in the adult brain. It is formed by the invagination of ependymal cells into the ventricles, which become richly vascularized.

Interstitial fluid

(ISF). The extracellular fluid filling the 'interstices' of the tissue, and bathing the cells.

Tight junction

A belt-like region of adhesion between adjacent cells. Tight junctions regulate paracellular flux, and contribute to the maintenance of cell polarity by stopping molecules from diffusing within the plane of the membrane.

Abluminal membrane

The endothelial cell membrane that faces away from the vessel lumen, towards the brain.

Meninges

The complex arrangement of three protective membranes surrounding the brain, with a thick outer connective tissue layer (dura) overlying the barrier layer (arachnoid), and finally the thin layer covering the glia limitans (pia). The sub-arachnoid layer has a sponge-like structure filled with CSF.

Circumventricular organs

(CVOs). Brain regions that have a rich vascular plexus with a specialized arrangement of blood vessels. The junctions between the capillary endothelial cells are not tight in the blood vessels of these regions, which allows the diffusion of large molecules. These organs include the organum vasculosum of the lamina terminalis, the subfornical organ, the median eminence and the area postrema.

Receptor-mediated transcytosis

The mechanism for vesicle-mediated transfer of substances across the cell, the first step of which requires specific binding of the ligand to a membrane receptor, followed by internalization (endocytosis).

Adsorptive-mediated transcytosis

The mechanism for vesicle-mediated transfer of substances across the cell, the first step of which involves nonspecific binding of the ligand to membrane surface charges, followed by internalization (endocytosis).

Adherens junction

A cell–cell junction also known as zonula adherens, which is characterized by the intracellular insertion of microfilaments. If intermediate filaments are inserted in lieu of microfilaments, the resulting junction is referred to as a desmosome.

Perivascular endfeet

The specialized foot-processes of perivascular astrocytes that are closely apposed to the outer surface of brain microvessels, and have specialized functions in inducing and regulating the BBB.

Pericyte

A cell of mesodermal origin, and contractile-phagocytic phenotype, associated with the outer surface of capillaries.

Orthogonal arrays of particles

(OAPs). The organized arrays (square lattice) of intramembranous particles detected by the freeze–fracture technique in certain astrocyte processes. First identified on the polarized endfeet on blood vessels and in the outer glial layer (glia limitans) below the pia, they have subsequently been shown to contain specific protein complexes held together by structural proteins.

Basal lamina

The extracellular matrix layer produced by the basal cell membrane, used as an anchoring and signalling site for cell–cell interactions.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Abbott, N., Rönnbäck, L. & Hansson, E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat Rev Neurosci 7, 41–53 (2006). https://doi.org/10.1038/nrn1824

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn1824

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing