Skip to main content

EDITORIAL article

Front. Physiol., 02 December 2021
Sec. Membrane Physiology and Membrane Biophysics
This article is part of the Research Topic Organization and Functional Properties of the Blood Brain Barrier View all 5 articles

Editorial: Organization and Functional Properties of the Blood-Brain Barrier

  • 1Discipline of Physiology and Biophysics, Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
  • 2Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute INCLIVA, University of Valencia, Valencia, Spain

Introduction

This Editorial provides a brief overview of the organization and functional properties of the blood-brain barrier, and introduces the foundational information in this book that contributes to these principles.

The Organizational Role of the Blood-Brain Barrier

The blood-brain barrier serves to restrict and control passage of solutes between the general circulation and brain extracellular fluid (Abbott and Friedman, 2012). Barrier function is due principally to the presence of specialized endothelial cells that line brain capillaries (Ge et al., 2005). These endothelial cells possess tight junctions that circumscribe the cells and largely prohibit extracellular movement of solutes between the cells (Hawkins and Davis, 2005; Tornavaca et al., 2015; Sweeney et al., 2019). The tight junctions demarcate two distinct plasmalemmal domains within the endothelium, including the luminal (blood facing) and abluminal (brain facing) plasma membranes (Sanchez del Pino et al., 1995; Peterson and Hawkins, 2003). Thus, transport across the blood-brain barrier is primarily transcellular, and net movement of solutes across the endothelium is determined by transport properties of the respective plasma membrane domains (Peterson and Hawkins, 1998, 2003; Hawkins et al., 2002). These substances may be altered by degradative enzymes associated with the barrier (el-Bacha and Minn, 1999), and barrier function is influenced by adjacent cells including astrocytes and pericytes that have led to the concept of a “neurovascular unit” (Hawkins and Davis, 2005; Abbott et al., 2006; Armulik et al., 2010; Sweeney et al., 2019).

Functions of the Blood-Brain Barrier

The blood-brain barrier contributes to homeostatic control of the central nervous system by modifying the volume and composition of brain extracellular fluid (Strange, 1992; Keep et al., 1998; Abbott and Friedman, 2012; Sweeney et al., 2019). Specific carrier proteins are present in both the luminal and abluminal plasma membranes of brain capillary endothelial cells that allow facilitated passive transport of nutrients from blood to brain, including glucose and amino acids (Drewes, 1998; Smith and Stoll, 1998; Hawkins et al., 2011; Peterson, 2019). Some amino acid transport systems possess sodium-dependent (i.e., secondary active) carriers in the abluminal membrane, thus providing possible mechanisms for regulation of transport (O'Kane et al., 1999; Peterson, 2019). Although the blood-brain barrier appears to be largely impermeable to peptides and proteins, there is evidence that some small peptides may enter the brain utilizing carrier-mediated processes (Banks, 2015; Sweeney et al., 2019). Furthermore, it appears that insulin and transferrin may cross the barrier by receptor-mediated endocytosis (Duffy and Pardridge, 1987; Pardridge et al., 1987). Unidirectional (blood-to-brain) fluid movement across the blood-brain barrier utilizes coordinated transport of salt and water that appears to be regulated (Strange, 1992; Keep et al., 1998; Peterson and Hawkins, 1998; Abbott and Friedman, 2012; Peterson, 2019; Sweeney et al., 2019). Sodium first enters passively into the cells utilizing carriers (e.g., Na/H antiporter, Na/K/Cl cotransporter, Na/Ca exchanger) in the luminal membrane of brain capillary endothelial cells. Intracellular sodium is then actively pumped out across the abluminal membrane by a Na/K-ATPase. It is generally believed that water passively follows sodium transport by utilizing water channels (i.e., aquaporin) in both the luminal and abluminal membranes (Nagelhus and Ottersen, 2013). There is evidence that fluid transport is regulated by centrally released peptide hormones (Strange, 1992; Abbott et al., 2006), and that normal fluid balance is achieved by its uptake and drainage into and out of the central nervous system.

Drug Delivery Across the Blood-Brain Barrier

One of the fundamental challenges to developing drugs that target the brain is the presence of the blood-brain barrier (Pardridge, 2012). However, understanding the mechanisms by which solutes are transported by blood-brain barrier endothelial cells provides a basis for designing drugs that are capable of traversing the barrier (Sweeney et al., 2019). For instance, pharmacological agents that are recognized by transport carriers that normally deliver nutrients to the brain would likely cross the blood-brain barrier. Another potential pathway involves utilization of normally occurring transcytotic pathways (Pardridge, 2012). Thus, coupling pharmacological agents that bind insulin or transferrin receptors has been shown to mediate drug delivery across the blood-brain barrier (Pardridge, 2012). In addition, procedures designed to loosen tight junctions have been used to promote intercellular movement of drugs across the barrier (Rapaport, 2000; Hsu et al., 2018). Nevertheless, each of these procedures for enabling passage of drugs across the blood-brain barrier is complicated by the presence of active efflux transporters in the endothelial cells that may limit or prevent net influx (Schinkel, 1999; Abbott et al., 2006; Reichel et al., 2011; Sweeney et al., 2019).

Alterations of the Blood-Brain Barrier in Disease

There is now evidence that dysfunction of the blood-brain barrier accompanies several diseases involving the central nervous system (Sweeney et al., 2019). For instance, blood-brain barrier dysfunction has been associated with Alzheimer's disease, amyotrophic lateral sclerosis, epilepsy, multiple sclerosis, Parkinson's disease, stroke, and traumatic brain injury (Papadopoulos et al., 2001; Lo et al., 2003; Marroni et al., 2003; Minagar and Alexander, 2003; Lee and Bendayan, 2004; Kortekaas et al., 2005; Peterson and Sukowski, 2019). Alterations of the barrier include changes in its permeability, transport properties, and regulatory mechanisms. Thus, understanding the normal structure and function of the blood-brain barrier, and defining the properties that are altered during pertinent neurological disorders, could provide a logical approach to designing effective therapeutics.

Book Chapters

The chapters in this book serve to: (1) review important discoveries in defining the organization and functional properties of the blood-brain barrier, and (2) introduce new concepts regarding its normal function or participation in disease processes.

The presentation by Partridge describes significant contributions of using brain microvessels as a model for investigating the blood-brain barrier and the neurovascular unit. Isolated brain microvessels consist of endothelial cells, pericytes, pre-capillary arteriolar smooth muscle cells, astrocyte foot processes, and nerve endings. They have been used as an in vitro model of the blood-brain barrier to: (1) produce cDNA libraries for genomic analyses, (2) quantify the presence of specific transporters and receptors using proteomics, (3) determine the cellular location of proteins expressed within the neurovascular unit by using immunolabeling, (4) study kinetic parameters of transport carriers, and (5) quantify dissociation constants of peptide binding involved in receptor-mediated transport.

The article by Locchead et al. focuses on functional properties of tight junctions in the blood-brain barrier, and how these are altered during certain pathological conditions. This presentation also describes how the blood-brain barrier may be manipulated therapeutically to allow for intercellular delivery of systemically administered drugs to the brain. Molecular elements of tight junctional function, regulation, and potential therapeutic manipulation are defined.

The paper by Brunner et al. describes a technique to quantify the contribution of claudins in the seal characterized by blood-brain barrier tight junctions. Xenopus lauvis oocytes are used as an expression system for claudins, and homophilic and heterophilic trans-interactions are characterized. The effect of hydrostatic pressure on the stability of cell-to-cell connections and their modulation are quantified as a function of claudin expression. This technique provides a potential basis for a more complete understanding of tight junction function and control in the blood-brain barrier.

The review by Zaragozå gives an inclusive over-view of amino acid transport by the blood-brain barrier. Both facilitative and secondary active transport processes are described, and the functional significance of a polarized distribution of amino acid transporters is discussed.

Author Contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of Interest

DP (inventor) and Rosalind Franklin University of Medicine and Science (assignee) have been awarded patents that are related to research done on treatment of stroke. They both may benefit financially upon commercialization of the patents. DP is a Professor Emeritus at Rosalind Franklin University of Medicine and Science and owns Harbor Biotechnology LLC, a company that seeks to commercialize the patents referenced above. Further details may be obtained from DP upon request.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abbott, N. J., and Friedman, A. (2012). Overview and introduction: the blood-brain barrier in health and disease. Epilepsia 53, 1–6. doi: 10.1111/j.1528-2012.03696.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Abbott, N. J., Ronnback, L., and Hansson, E. (2006). Astrocyte-endothelial interactions at the blood-brain barrier. Nat. Rev. Neurosci. 7, 41–53. doi: 10.1038/nrn1824

PubMed Abstract | CrossRef Full Text | Google Scholar

Armulik, A., Genove, G., Mae, M., Nisancioglu, M. H., Wallgard, E., and Niaudet, C. (2010). Pericytes regulate the blood-brain barrier. Nature 468, 557–561. doi: 10.1038/nature09522

PubMed Abstract | CrossRef Full Text | Google Scholar

Banks, W. A. (2015). Peptides and the blood-brain barrier. Peptides 72, 16–19. doi: 10.1016/j.peptides.03.010

CrossRef Full Text | Google Scholar

Drewes, L. R. (1998). “Biology of the blood–brain glucose transporter,” in Introduction to the Blood–Brain Barrier, ed W. M. Pardridge (Cambridge: Cambridge University Press), 165–174. doi: 10.1017./CBO9780511570216.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Duffy, K. R., and Pardridge, W. M. (1987). Blood-brain barrier transcytosis of insulin in developing rabbits. Brain Res. 420, 32–38. doi: 10.1016/0006-8993(87)90236-8

PubMed Abstract | CrossRef Full Text | Google Scholar

el-Bacha, R., and Minn, S. (1999). Drug metabolizing enzymes in cerebrovascular endothelial cells afford a metabolic protection to the brain. Cell. Mol. Biol. 45, 15–25.

PubMed Abstract | Google Scholar

Ge, S., Song, L., and Pachter, J. (2005). Where is the blood-brain barrier…really? J. Neurosci. Res. 79, 421–427. doi: 10.1002/jnr.20313

PubMed Abstract | CrossRef Full Text | Google Scholar

Hawkins, B. T., and Davis, T. P. (2005). The blood-brain barrier/neurovascular unit in health and disease. Pharmacol. Rev. 57, 173–185. doi: 10.1124/pr.57.2.4

PubMed Abstract | CrossRef Full Text | Google Scholar

Hawkins, R. A., Peterson, D. R., and Vina, J. R. (2002). The complementary membranes forming the blood–brain barrier. Int. Union Biochem. Mol. Biol. Life 54, 101–108. doi: 10.1080/15216540214541

PubMed Abstract | CrossRef Full Text | Google Scholar

Hawkins, R. A., Viña, J. R., Peterson, D. R., O'Kane, R., Mokashi, A., and Simpson, I. A. (2011). “Amino acid transport across each side of the blood-brain barrier,” in Amino Acids in Nutrition and Health, ed J. P. F. D'Mello (Oxford: CABI), 191–214. doi: 10.1079./9781845937980.0191

CrossRef Full Text | Google Scholar

Hsu, P. H., Lin, Y. T., Chung, Y. H., Lin, K. J., Yang, L.Y., and Yen, T. C. (2018). Focused ultrasound-induced blood-brain barrier opening enhances GSK-3 inhibitor delivery for amyloid-beta plaque reduction. Sci. Rep. 8, 12882–12890. doi: 10.1038/s41598-018-31071-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Keep, R., Ennis, S., and Betz, A. (1998). “Blood-brain barrier ion transport,” in Introduction to the Blood–Brain Barrier, ed W. M. Pardridge (Cambridge: Cambridge University Press), 207–213. doi: 10.1017/CBO9780511570216.022

PubMed Abstract | CrossRef Full Text

Kortekaas, R., Leenders, K. L., van Oostrom, J. C., Vaalburg, W., Bart, J., and Willemsen, A. T. (2005). Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol. 57, 176–179. doi: 10.1002/ana.20369

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, G., and Bendayan, R. (2004). 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. doi: 10.1023/B:PHAM.0000036905.82914.8e

PubMed Abstract | CrossRef Full Text | Google Scholar

Lo, E. H., Dalkara, T., and Moskowitz, M. A. (2003). Mechanisms, challenges and opportunities in stroke. Nat. Rev. 4, 399–415. doi: 10.1038/nrn1106

PubMed Abstract | CrossRef Full Text | Google Scholar

Maroni, M., Marchi, N., Cucullo, L., Abbott, N. J., Signorelli, K., and Janigro, D. (2003). Vascular and parenchymal mechanisms in multiple drug resistance: a lesson from human epilepsy. Curr. Drug Targets 4, 297–304. doi: 10.2174/1389450033491109

PubMed Abstract | CrossRef Full Text | Google Scholar

Minagar, A., and Alexander, J. S. (2003). Blood-brain barrier disruption in multiple sclerosis. Multiple Sclerosis 9, 540–549. doi: 10.1191/1352458503ms965oa

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagelhus, E. A., and Ottersen, O. P. (2013). Physiological roles of aquaporin-4 in brain. Physiol. Rev. 93, 1543–1562. doi: 10.1152/physrev.00011.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

O'Kane, R. L., Martínez-López, M. R., DeJoseph, M. R., Viña, J. R., and Hawkins, R. A. (1999). Na(+)-dependent glutamate transporters (EAAT1, EAAT2, and EAAT3) of the blood-brain barrier. A mechanism for glutamate removal. J. Biol. Chem. 274, 31891–31895. doi: 10.1074/jbc.274.45.31891

PubMed Abstract | CrossRef Full Text | Google Scholar

Papadopoulos, M. C., Saadoun, S., Davies, D. C., and Bell, B. A. (2001). Emerging molecular mechanisms of brain tumour oedema. Br. J. Neurosurg. 15, 1–108. doi: 10.1080/02688690120036775

PubMed Abstract | CrossRef Full Text | Google Scholar

Pardridge, W. M. (2012). Drug transport across the blood-brain barrier. J. Cereb. Blood Flow Metab. 32, 1959–1972. doi: 10.1038/jcbfm.2012.126

PubMed Abstract | CrossRef Full Text | Google Scholar

Pardridge, W. M., Eisenberg, J., and Yang, J. (1987). Human blood-brain barrier transferrin receptor. Metab. Clin. Exp. 36, 892–895. doi: 10.1016/0026-0495(87)90099-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Peterson, D. R. (2019). Blood-Brain Barrier. Chichester: eLS. John Wiley and Sons, Ltd. doi: 10.1002/9780470015902.a0000023.pub4

PubMed Abstract | CrossRef Full Text

Peterson, D. R., and Hawkins, R. A. (1998). “Isolation and behavior of plasma membrane vesicles made from cerebral capillary endothelial cells,” in Introduction to the Blood–Brain Barrier, W. M. Pardridge (Cambridge: Cambridge University Press), 62–70. doi: 10.1017/CBO9780511570216.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Peterson, D. R., and Hawkins, R. A. (2003). “Transport studies using membrane vesicles,” in Blood-Brain Barrier: Biology and Protocols, ed S. Nag (Totowa, NJ: The Humana Press), 233–248. doi: 10.1385/1-59259-419-0:233

PubMed Abstract | CrossRef Full Text | Google Scholar

Peterson, D. R., and Sukowski, E. J. (2019). “Prevention of oxidative injury associated with thrombolysis for ischemic stroke,” in Antioxidants, ed E. Shalaby (London: IntechOpen), 197–127. doi: 10.5772/intechopen.84774

PubMed Abstract | CrossRef Full Text | Google Scholar

Rapaport, S. I. (2000). Osmotic opening of the blood-brain barrier: principles, mechanism, and therapeutic application. Cell. Mol. Neurol. 20, 217–230. doi: 10.1023/A:1007049806660

PubMed Abstract | CrossRef Full Text | Google Scholar

Reichel, V., Burghard, S., and Huber, J. (2011). P-glycoprotein and breast cancer resistance protein expression and function at the blood-brain barrier and blood-cerebrospinal fluid barrier (choroid plexus) in streptozotocin-induced diabetes in rats. Brain Res. 1370, 238–245. doi: 10.1016/j.brainres.11.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez del Pino, M. M., Hawkins, R. A., and Peterson, D. R. (1995). Biochemical discrimination between luminal and abluminal enzyme and transport activities of the blood-brain barrier. J. Biol. Chem. 270, 14907–14912. doi: 10.1074/jbc.270.25.14907

PubMed Abstract | CrossRef Full Text | Google Scholar

Schinkel, A. (1999). P-glycoprotein, a gatekeeper in the blood-brain barrier. Adv. Drug Delivery Res. 36, 179–194. doi: 10.1016/S0169-409X(98)00085-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, Q. R., and Stoll, J. (1998). “Blood–brain barrier amino acid transport,” in Introduction to the Blood–Brain Barrier, ed W. M. Pardridge (Cambridge: Cambridge University Press), 188–197. doi: 10.1017/CBO9780511570216.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Strange, K. (1992). Regulation of solute and water balance and cell volume in the central nervous system. J. Am. Soc. Nephrol. 3, 12–27. doi: 10.1681/ASN.V3112

PubMed Abstract | CrossRef Full Text | Google Scholar

Sweeney, M. D., Zhao, Z., and Montagne, A. (2019). Blood-brain barrier: from physiology to disease and back. Physiol. Rev. 99, 21–78. doi: 10.1152/physrev.00050.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Tornavaca, O., Chia, M., Dufton, N., Brils, G., Cuevas, A., and Hespel, T. (2015). ZO-1 controls endothelial adherens junctions, cell-cell tension, angiogenesis, and barrierformation. J. Cell Biol. 208, 821–838. doi: 10.1083/jcb.201404140

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: structure, transport, drug delivery, disease, blood-brain barrier

Citation: Peterson DR, Hawkins RA and Viña JR (2021) Editorial: Organization and Functional Properties of the Blood-Brain Barrier. Front. Physiol. 12:796030. doi: 10.3389/fphys.2021.796030

Received: 15 October 2021; Accepted: 10 November 2021;
Published: 02 December 2021.

Edited and reviewed by: Christoph Fahlke, Helmholtz-Verband Deutscher Forschungszentren (HZ), Germany

Copyright © 2021 Peterson, Hawkins and Viña. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Darryl R. Peterson, darryl.peterson@rosalindfranklin.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.