The FEBS Journal - 2015 - Keaney - The Dynamic Blood Brain Barrier

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

STATE-OF-THE-ART REVIEW

The dynamic blood–brain barrier


James Keaney and Matthew Campbell
Smurfit Institute of Genetics, Trinity College Dublin, Ireland

Keywords With the endothelium as its central unit, the blood–brain barrier (BBB) is
Alzheimer’s disease; astrocytes; blood–brain a complex multicellular structure separating the central nervous system
barrier; GLUT-1; gut microbiota; Mfsd2a;
(CNS) from the systemic circulation. Disruption of the BBB has now been
neurovascular unit; pericytes; tight
implicated in a multitude of acute and chronic CNS disorders indicating
junctions; transcytosis
the potentially devastating effects of BBB breakdown on brain function.
Correspondence However, the healthy BBB is not an impermeable wall, but rather a com-
M. Campbell, Smurfit Institute of Genetics, munication ‘centre’, responding to and passing signals between the CNS
Trinity College Dublin, Dublin 2, Ireland and blood. New studies are identifying BBB-specific transport pathways
Fax: +353 189 638 48 that tightly regulate the entry and exit of molecules to and from the brain.
Tel: +353 189 624 84
They are revealing a highly plastic barrier in which dynamic changes in
E-mail: Matthew.Campbell@tcd.ie
BBB components like paracellular tight junction complexes can contribute
(Received 29 May 2015, revised 20 July to BBB maintenance. Here, we provide a succinct overview of the current
2015, accepted 12 August 2015) state-of-play in BBB research and summarize novel findings into BBB
regulation in homeostatic regulation of the brain.
doi:10.1111/febs.13412

Introduction
In the mammalian body, numerous biological barriers properties distinct from endothelial cells in other tis-
separate the circulating blood from the interstitial fluid sues: the presence of BBB-specific transporter and
that surrounds tissues. Positioned along the blood ves- receptor proteins to control entry and exit of metabo-
sels of the central nervous system (CNS), the blood– lites across cells (transcellular transport) and high
brain barrier (BBB) is perhaps the most selective and electrical resistance tight junctions (TJs) to limit move-
tightly controlled of these barriers, reflecting the ment between adjacent cells (paracellular transport);
brain’s critical roles in cognition, regulating metabo- and low levels of transcytotic vesicles compared to
lism and co-ordinating the functions of peripheral peripheral endothelia and an absence of fenestrae
organs. Because communicating this information (small pores that allow rapid passage of molecules in
depends on fine control of electrical and chemical sig- peripheral endothelial cells) [1,2].
nals between neurons, the brain requires a precise and Although the endothelial cells of the CNS vascula-
balanced microenvironment. The BBB is therefore ture act as the principal barrier unit, it is now recog-
essential in regulating the influx and efflux of ions, nized that an intricate network of molecular crosstalk
oxygen and nutrients between the blood and brain among a variety of cell types is required to form the
compartments and protecting the delicate neural tissue BBB during embryonic development and to maintain
by restricting the invasion of toxins and pathogens. To its integrity in adulthood [3]. Cell–cell interactions
sustain this robust barrier, CNS endothelial cells have within the ‘neurovascular unit’ (NVU) – the milieu of

Abbreviations
Ab, amyloid-b; AD, Alzheimer’s disease; AJ, adherens junctions; BBB, blood–brain barrier; CNS, central nervous system; CSF, cerebrospinal
fluid; LRP1, lipoprotein receptor-related protein 1; NVU, neurovascular unit; RAGE, receptor for advanced glycation end products; TBI,
traumatic brain injury; TEM, transendothelial leukocyte migration; TJ, tight junction; VE, vascular endothelial; WNV, West Nile virus; ZO-1,
zonula occludens-1.

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4067


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

neurons, astrocytes, pericytes, microglia and other the grafted tissue developed endothelial cells with
components of the brain parenchyma that communi- characteristics of the nonleaky BBB such as TJs and
cate with endothelial cells – are helping to explain how minimal transcytotic vesicles. When dorsal mesoderm
the unique characteristics of the BBB are induced and from embryonic quails was transplanted into the embry-
how disease-associated dysfunction of any one NVU onic chick brain, the vessels developing in the grafted
component can impact BBB permeability [4]. Recent tissue lacked barrier properties [10]. This suggested that
work has also identified how deficiencies in specific interactions between the nascent vascular tissue and the
macromolecule transporters (e.g. GLUT-1, Mfsd2a) surrounding neural microenvironment are crucial to
[5–7] and paracellular TJ complexes [8] at the BBB can BBB development. Much attention has since focused on
affect overall cerebrovascular integrity. This has impli- the roles of two cell types, astrocytes and pericytes,
cations in the study of CNS disorders like Alzheimer’s which exist in close anatomical association with brain
disease (AD) and in developing novel drug-delivery endothelial cells. Along with neurons, microglia (the
systems based on BBB biology. Insights from studies brain’s resident macrophages), peripheral immune cells,
of infection and innate immunity in the CNS are also the noncellular basement membrane and other compo-
challenging the oft-used analogy of the BBB as a nents, it has become clear that no cell type in the brain
‘brick wall’ guarding the precious neuronal tissue [9]. functions in isolation, but instead is in constant commu-
There is an increased awareness that dynamic changes nication with other NVU members. See Fig. 1 for sche-
in the levels and cellular distribution of BBB compo- matic representation of the BBB.
nents such as TJ proteins help to maintain a healthy
and robust BBB under physiological conditions.
Astrocytes
Indeed, we discuss the emerging influences of diverse
biological processes like sleep–wake behaviour and gut Astrocytes are glial cells that help support and protect
microbiota on BBB development and integrity, high- neurons by controlling neurotransmitter and ion con-
lighting a complex interplay between CNS- and centrations to maintain the homeostatic balance of the
peripheral-derived signals converging at the BBB on a neural microenvironment, by modulating synaptic
daily basis. transmission and by regulating immune reactions [11].
Astrocytes are also known to interact with endothelial
cells through their end-feet projections that encircle the
 lange of
The ‘neurovascular unit’: a me
abluminal side of cerebral capillaries [12]. In the adult
cell types
brain, such interactions are important in synchronizing
A dominant question in early BBB research concerned metabolite levels with cerebral blood flow and vasodi-
whether the unique properties of cerebral endothelial lation, and regulating brain water content [13]. For
cells were intrinsic to this cell type or whether signals example, the most abundant water channel protein,
from the surrounding brain microenvironment induced aquaporin 4, is predominantly expressed in astrocytic
BBB formation. In early transplantation experiments by end-feet surrounding CNS vessels [14]. Astrocytes are
Stewart and Wiley [10] using quail chick chimeras, it also a key cellular support of BBB integrity. Recent
was found that following grafting of immature avascu- in vitro and in vivo molecular studies have revealed
lar brain tissue from embryonic quails into the coelom several effector molecules released by astrocytes that
of chick embryos, the abdominal vessels vascularizing function to enhance and maintain barrier tightness,

Fig. 1. The neurovascular unit comprises


endothelial cells, pericytes and astrocytes
that together confer unique properties on
the blood–brain barrier. The endothelial
cells that line the cerebral vasculature
have numerous transcellular transporters
in addition to well-evolved tight junctions
that limit the passive paracellular diffusion
of all but the smallest of solutes and ions.

4068 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

primarily members of the Hedgehog family [15], the nascent vessels of the embryonic brain (driven by
renin–angiotensin hormone system [16], and the vascular endothelial growth factor and Wnt/b–catenin
cholesterol and phospholipid transporter molecule modelling among other pathways) [3,27–32], pericyte
apolipoprotein E [17]. Release of apolipoprotein E recruitment is crucial to establishing BBB characteris-
from astrocytes, for example, regulates endothelial TJs tics. Complete loss of pericytes in platelet-derived
by signalling through the low-density lipoprotein recep- growth factor (Pdgfb) or Pdgfrb knockout mice has
tor-related protein 1 (LRP1) on both pericytes and been shown to result in CNS microhaemorrhages, dys-
endothelial cells of CNS microvessels [17,18]. functional TJs, increased vascular permeability and
The role of astrocytes in BBB development in the embryonic lethality [23,33]. It is thought that PDGFb
immature brain is more controversial. When Janzer release by endothelial cells of sprouting vessels pro-
and Raff [19] injected neonatal rat astrocytes into the motes the recruitment and proliferation of PDGFRb-
anterior chamber of the eye, only vessels formed in the positive pericytes, which in turn decrease transendothe-
presence of astrocytes excluded Evans blue dye (in- lial trafficking and inhibit the expression of molecules
jected systemically). Based on these results, it was pro- in endothelial cells that promote immune cell infiltra-
posed that astrocytes were necessary for the formation tion [4,23]. Following pericyte recruitment to develop-
of impermeable TJs, but in a follow-up study by a sep- ing vessels, further communication between pericytes
arate group, no ultrastructural changes in the develop- and neighbouring endothelial cells is established via
ing vessels of the astrocyte grafts was observed [20]. transforming growth factor b and its receptor
In vitro co-cultures of endothelial cells with astrocytes (TGFRb) [34]. Importantly, these pericyte effects on
or astrocyte-conditioned media have been shown to BBB formation precede astrocyte differentiation by
induce more complex TJs, elevated expression of trans- one week [23].
porters and increased transendothelial electrical resis- Pericytes are also vital to BBB integrity during
tance although these studies are limited by the use of adulthood because Pdgfrb knockout mice exhibit age-
adult endothelial cells [21,22]. Furthermore, the finding dependent BBB dysfunction as a result of reduced TJ
that astrocytes are not present in the developing brain protein expression [35], whereas young adult mice with
during the time of initial vascularization lends support hypomorphic alleles of Pdgfb display defects in BBB
to a role for astrocytes in TJ maintenance and not for- integrity as a result of increased rates of endothelial
mation [23]. More recently, however, production and transcytosis [36]. Intriguingly, Armulik and colleagues
release of retinoic acid by astrocyte precursors (radial [36] also found that pericytes not only regulate mature
glial cells) has been shown to act through the retinoic BBB integrity, but also function to guide astrocytic
acid receptor-b on developing brain endothelial cells to foot processes to cerebral vessel walls and mediate the
induce BBB properties [24]. polarization of astrocytic end-feet, again emphasizing
the complex interdependency among components of
the NVU. Another recent study has highlighted the
Pericytes
involvement of pericytes in disease phenotypes such as
Although astrocyte–endothelial interactions along neurodegeneration. Progressive loss of pericytes in a
mature vessels are essential for BBB integrity and may mouse model of AD lacking a single Pdgfrb allele
also help seal the barrier in the final stages of BBB (APPSw/Pdgfrb+/ ) accelerated the hallmarks of AD
formation, pericytes have emerged as a central NVU pathology including elevated brain amyloid b (Ab)
component in the first stages of ‘barriergenesis’. Peri- levels, tau pathology and early neuronal loss [37].
cytes are perivascular cells of mesodermal origin whose These combined findings underscore the importance of
elongated processes ensheath CNS vessel walls. The pericytes not only in barrier differentiation during
CNS vasculature has significantly higher pericyte cov- embryonic development, but also in maintaining a
erage compared with peripheral vessels and pericytes healthy and robust BBB and proper neural function in
have an important role in regulating capillary diame- adulthood.
ter, cerebral blood flow and extracellular matrix pro-
tein secretion and levels [25]. Indeed, a recent study
Microglia
has highlighted an essential role of capillary pericytes
in regulating cerebral blood flow – the neurotransmit- Derived from haematopoietic precursors that migrate
ter glutamate promotes the release of messengers from the yolk sac into the CNS parenchyma, microglia
including prostaglandin E2 and nitric oxide that help act as the brain’s main line of defence past the BBB
dilate capillaries by actively relaxing pericytes [26]. and play a vital role in innate immune responses in the
During early angiogenesis and barrier formation along CNS [38]. Although located in close proximity to

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4069


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

endothelial cells of brain microvessels, little is known structural proteins secreted by astrocytes, pericytes and
about potential microglial–endothelial communication endothelial cells. The basement membrane helps to
in forming and regulating the homeostatic BBB. One regulate crosstalk between NVU components and to
study has demonstrated that microglia associate with this end, Yao and colleagues [48] have recently shown
endothelial tip cells along nascent vessels in the devel- that a brain-specific basement membrane component,
oping brain and promote the fusion of tip cells in the astrocytic-derived laminin, helps to regulate pericyte
stages following vascular endothelial growth factor- differentiation via binding to the integrin a2 receptor
mediated tip cell induction [39]. At present, more is on pericytes. Disruption of this basement membrane
known about the activation of microglia in CNS disor- protein in the brain converts pericytes from a resting
ders like AD and multiple sclerosis, which are associ- to a contractile phenotype, which in turn decreases
ated with BBB breakdown and neuroinflammation. In aquaporin 4 levels in astrocytic end-feet and decreases
these conditions, microglial activation may be both a endothelial TJ expression [48].
cause and consequence of BBB dysfunction [40]. It is now clear that molecular communication
Microglia can exist in one of two active states: in the between cellular and noncellular members of the
classically activated M1 pathway, microglia primarily NVU, first established during early CNS angiogenesis
release proinflammatory cytokines like interleukin-1b and barriergenesis, must remain in place to ensure
and tumour necrosis factor-a, whereas in the alterna- BBB tightness in adulthood. Moreover, it suggests that
tive M2 pathway microglia are involved in tissue if this fine balance is disrupted, it may directly or indi-
repair, phagocytosing damaged neurons and foreign rectly contribute to CNS disease pathogenesis. As we
material, releasing chemokines and vascular endothe- see in the next sections, these NVU components help
lial growth factor, and activating neurotrophic path- to establish and maintain transport pathways located
ways [41]. In multiple sclerosis, the release of across brain endothelial cells, the breakdown of which
proinflammatory cytokines and reactive oxygen species can lead to uncontrolled BBB permeability.
by activated microglia can exacerbate myelin damage
and increase BBB permeability potentially via down-
BBB transport routes in development
regulation of adherens junction [AJ, such as vascular
and disease
endothelial (VE)-cadherin] and TJ (occludin and clau-
din-5) proteins [42,43]. In AD, T-cell transmigration Passive diffusion across brain endothelia is limited to
across the BBB can also be driven by microglia- lipid-soluble molecules of approximately < 180 Da in
derived signals such as tumour necrosis factor-a [44]. size and with fewer than 10 hydrogen bonds [49].
Tumour necrosis factor-a secretion by microglia in Instead, distinct BBB transcellular transport systems
response to Ab leads to upregulation of MHC class I exist to tightly regulate the CNS entry and exit of
in brain endothelial cells, which in turn provides a hydrophilic molecules. Carrier-mediated transport is
mechanism for T-cell migration into the brain [44]. the major route for essential nutrients like glucose,
Recruitment of these peripheral lymphocytes may help fatty acids and amino acids to travel across the BBB
or hinder resident microglia in resolving brain injury [1]. New studies are revealing BBB-enriched compo-
depending on the CNS disease and the disease stage nents that lead ‘double lives’ – not only as trans-
[9]. Brain endothelial cells can also secrete molecules porters of essential molecules, but also as important
that cause microglial activation. Microvessels from regulators of BBB permeability. In the context of
AD patients have been found to release high levels of CNS disease, we also consider the BBB transcytosis
the neurotoxic protease thrombin [45]. Previous work of Ab – the major pathogenic component in Alzhei-
has highlighted how thrombin can increase nitric oxide mer’s disease.
levels, activate microglia and contribute to the loss of
dopaminergic neurons [46]. Along with bone-marrow-
GLUT-1
derived perivascular macrophages that can act as anti-
gen-presenting cells [47], a complex interplay between The glucose transporter GLUT-1 (encoded by the
systemic- and CNS-derived immune cells exists at the Slc2a1 gene) is enriched in CNS endothelial cells com-
BBB. pared with peripheral endothelia [50]. The importance
of GLUT-1 in brain glucose metabolism is reflected in
human GLUT-1 deficiency syndromes in which
Basement membrane
affected individuals suffer from infantile seizures, cog-
An often-overlooked component of the NVU is the nitive impairments and developmental delay [51]. Fur-
basement membrane – the extracellular matrix of thermore, GLUT-1 deficiency has profound impacts

4070 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

on BBB physiology and neurovascular function in embryonic and adult stages as a result of increased
because Slc2a1+/ mice have been found to have endothelial vesicular transcytosis [7]. Similar to
decreased brain glucose uptake, reduced cerebral blood GLUT-1, this suggests a double function for Mfsd2a
flow and increased BBB permeability due to lower in mediating nutrient supply from the blood into the
TJ protein levels [5]. This suggests that loss of BBB- brain while also regulating BBB integrity. Further-
specific transporters can ultimately impact general more, because pericyte-deficient mice display reduced
BBB integrity and also has implications in neurodegen- endothelial Mfsd2a levels and increased vesicular traf-
erative disorders. GLUT-1 levels are known to be ficking is seen in both pericyte-deficient and Mfsd2a
decreased in brain microvessels of AD patients [52] knockout mice, this suggests that one way in which
and correlate with reduced brain sugar uptake and pericytes support BBB transport pathways and BBB
subsequent cognitive decline [53]. It is still unclear integrity is via the control of Mfsd2a expression [7].
whether reduced GLUT-1 levels in AD represent lower Determining whether individuals with mutant Mfsd2a-
energy demands in a degenerating brain or whether associated microcephaly syndromes display BBB
the primary insult occurs at the BBB and precedes breakdown in addition to deficient lysophosphatidyl-
brain atrophy. However, PET imaging studies have choline transport will be of great interest.
suggested that altered glucose transport occurs early in
AD [54,55]. Analysis of phenotypes in APPSw/
Ab transcytosis
Slc2a1+/ mice, an AD mouse model with deficits in
GLUT-1, indicates that BBB breakdown following Both brain-derived and peripheral Ab is transported
reduced GLUT-1 precedes and exacerbates features of across the BBB via receptor-mediated transcytosis.
AD pathology including reduced Ab clearance across Based on recent measurements of venous to arterial
the BBB, elevated brain Ab levels, impaired neural Ab concentration ratios in humans, ~ 25% of Ab
activity and neuronal loss [5]. In light of these findings, clearance from the brain occurs at the BBB [58].
studying perturbations of BBB transporters in the Highly expressed in brain endothelial cells of the BBB,
early stages of other CNS disorders may yield novel the ATP-binding cassette efflux transporter P-glyco-
insights into disease mechanisms. protein (Pgp) has been implicated in Ab transport out
of the brain as APP/Pgp-null transgenic mice show
increased brain Ab levels [59,60]. However much atten-
Mfsd2a
tion has focused on the receptor for advanced glyca-
Mfsd2a has been recently identified as a critical com- tion end products (RAGE) and low-density LRP1 as
ponent of BBB formation and integrity. Mfsd2a is a the central Ab transporters at the BBB. Elucidating
brain endothelium-enriched receptor for the omega 3 these transcellular pathways of Ab movement has
fatty acid, docosahexaenoic acid (transported in the revealed how disrupting their normal activity can
form of lysophosphatidylcholine), which is essential accelerate Ab build up and AD pathogenesis. RAGE
for normal brain growth and function [6]. Two human is an Ab receptor found on the surface of neurons and
genetic analyses have recently identified missense microglia and is also the major endothelial receptor
mutations in the Mfsd2a gene that lead to autosomal for Ab influx from the blood to the brain [61]. RAGE
recessively inherited microcephaly syndromes [56,57]. expression at the BBB is increased in the brain
Interestingly, all three Mfsd2a mutations identified in endothelium of many AD patients as well as in AD
these studies did not affect Mfsd2a protein expression transgenic mouse models, likely due to accumulation
or cell surface localization. Instead, the Mfsd2a variant of its ligand Ab [61]. LRP1 receptor mediates cellular
affecting individuals in one pedigree (encoding uptake and internalization of Ab for lysosomal degra-
p.Ser339Leu) leads to partial inactivation of Mfsd2a’s dation in neurons, astrocytes, microglia and vascular
lysophosphatidylcholine transport activity and non- smooth muscle cells [62,63]. Working in parallel with
lethal microcephaly and intellectual disability pheno- RAGE, LRP1 is a major Ab efflux transporter and
types [56]. Two other Mfsd2a variants discovered in LRP1/Ab binding at the abluminal side of the BBB
separate families (p.Thr159Met and p.Ser166Leu) mediates Ab clearance from brain to blood [64].
cause complete Mfsd2a inactivation and these individ- Decreased LRP1 levels have been detected in brain
uals present with severe microcephaly, ventricu- microvessels in aged rodents, nonhuman primates and
lomegaly (enlarged lateral ventricles), and seizures that humans, as well as in AD mouse models and AD
proves fatal in the early years of life [57]. In rodents, patients [64,65]. More recently, the Zlokovic group
Mfsd2a knockout mice have reduced brain levels of have identified PICALM as being involved in the
docosahexaenoic acid [6] but also display a leaky BBB internalization and trafficking of LRP1/Ab complexes

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4071


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

across the BBB [66]. PICALM was previously identi- ulate paracellular permeability [8]. Loss of AJ proteins
fied from genome-wide association studies as a genetic like VE-cadherin has been reported in brain microves-
risk factor in AD [67]. sels of multiple sclerosis patients [79], although more
research is needed to understand the involvement of
AJs in CNS disease pathology. TJ breakdown has
Paracellular multiprotein complexes: adherens
been linked to increased BBB permeability and neu-
junctions and tight junctions
ronal dysfunction in chronic neurological disorders
Located between neighbouring brain endothelial cells, like amyotrophic lateral sclerosis and AD [80–82].
adherens junctions (AJs) and TJs are distinct multipro- However, detailed analyses of events at the BBB fol-
tein complexes that interconnect at a structural and lowing ischaemic stroke and traumatic brain injury
functional level to seal the paracellular gap. Cadherins (TBI) reveal highly dynamic and kinetic rearrange-
are the major protein family of AJs and include VE- ments of TJ complexes and other pathways in the min-
cadherin, which forms homophilic interactions across utes, hours and days after injury. Along with recent
adjacent endothelial cells [68]. Although the roles of studies of infection and innate immunity in the CNS,
AJs specifically at the BBB are not fully resolved, AJs this is helping to consider TJ complexes and the BBB
are believed to initiate cell–cell contacts, promote as adaptive structures that can quickly respond to sig-
endothelial cell survival, establish cell polarity and nals emanating from either the brain or the blood.
respond to stimuli via interactions with the catenin
complex and actin cytoskeleton [69,70]. By contrast to
Dynamic BBB remodelling: the flexible
AJs that are present in all vascular beds of the mam-
TJ
malian body, high electrical resistance continuous TJs
are specific to the brain vasculature. TJ protein com-
Stroke and TBI
plexes play a critical role in neuronal homeostasis by
restricting the paracellular diffusion of ions and In rodent studies, both ischaemic stroke and TBI are
macromolecules across the BBB and by linking extra- characterized by a ‘biphasic’ opening of the BBB fol-
cellular stimuli with intracellular signalling in the brain lowing the initial insult. Although reperfusion of the
endothelium. Major TJ proteins at the BBB include a infarct site after an ischaemic stroke is necessary to
number of interacting transmembrane components: support damaged neuronal tissue, return of the blood
occludin, claudins (in particular, claudin-5 and clau- supply can also lead to further neuronal damage and
din-12) and members of the junctional adhesion mole- cerebrovascular injury [83]. In the transient middle
cule family (e.g. JAM-4) [71,72]. Anchored to these cerebral artery occlusion model of ischaemic stroke, a
transmembrane TJ proteins, intracellular components biphasic increase in BBB permeability during reperfu-
such as zonula occludens-1 (ZO-1) act as a scaffold sion (at ~ 6 h and 48–72 h) corresponds to increased
connecting TJ strands with the actin cytoskeleton of vasogenic oedema formation [84]. In an elegant study
brain endothelial cells [73]. Where three brain endothe- using fluorescent tracers and two-photon time-lapse
lial cells meet, tricellulin and lipolysis-stimulated imaging in the transient middle cerebral artery occlu-
lipoprotein receptor have been identified as potential sion model, Knowland and colleagues [85] have
regulators of paracellular permeability at tricellular recently mapped the dynamic events at the BBB that
contacts [74,75]. lead to barrier deficits in the hours after stroke. Using
Increasing evidence of crosstalk between compo- Claudin-5:eGFP transgenic mice to visualize endothe-
nents of both AJs and TJs highlights a sophisticated lial TJs, ultrastructural changes in TJ assemblies such
system in establishing and regulating the paracellular as gaps (discontinuous TJs) and protrusions (exten-
barrier. ZO-1, for example, controls VE-cadherin- sions from linear TJ strands), and decreased occludin
mediated cell–cell tension and cytoskeletal organization and ZO-1 staining were found 48–58 h after stroke
[76]. Conversely, VE-cadherin can upregulate claudin-5 and correlated with delayed leakage of biocytin-TMR
and promote its junctional localization [77]. In and IgG [85]. At early time-points, however, there
response to shear stress, VE-cadherin can also transmit were minimal deficits in TJ integrity, instead increased
signals via Tiam1/Rac1, which reduces levels of tyro- rates of endocytosis and transcytosis (measured by
sine-phosphorylated occludin and increases barrier uptake and transport of fluorescently labelled albumin)
strength [78]. In addition to transcriptional up- and were detected as early as 6 h after transient middle
downregulation and changes in cellular localization, cerebral artery occlusion [85]. This distinct sequence of
post-translational phosphorylation of junctional pro- events at the BBB – increased transcytosis followed by
teins at serine, threonine and tyrosine residues can reg- TJ remodelling – highlights the dynamic involvement

4072 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

of both transcellular and paracellular pathways in served during this process. This may be due to
modulating BBB permeability in the hours and days PECAM-1-mediated recruitment of the lateral border
following a cerebral infarction. TJ plasticity and recycling compartment, a vesicular-membrane com-
biphasic opening of the BBB are also observed in the partment, to the TJ that can laterally displace TJ com-
penumbral region or neural tissue surrounding the site ponents while also sealing the intercellular gap [90].
of a TBI. In a cortical cold-induced injury model of Although chronic BBB breakdown and leukocyte infil-
TBI, TJ protein levels are altered post injury [86] and tration in conditions such as multiple sclerosis is likely
BBB permeability is evident in the penumbra at 24 detrimental [91], bone marrow-derived microglia are
and 72 h, but not 48 h after injury [87]. Interestingly, known to be crucial for restricting senile plaque for-
we have found that short interfering RNA-mediated mation in AD [92]. Recruitment of T cells to the CNS
suppression of claudin-5 in this model enhances move- can also contribute to hippocampal neurogenesis and
ment of water from brain to blood, reducing cerebral spatial learning in adulthood [93], thus the BBB likely
oedema and improving cognitive function in rodent plays an active role in mediating contact between the
models [87]. Although BBB breakdown in the TBI CNS and peripheral immune system.
region likely exacerbates neuronal dysfunction and The paracellular pathway of the BBB can also be
inflammation, TJ complexes in the penumbra may hijacked by pathogens like bacteria and viruses to
adapt to this fluid accumulation to alleviate swelling invade the CNS. Bacterial meningitis in newborns is
and protect the surrounding neuronal tissue. commonly caused by group B Streptococcus, a Gram-
positive bacteria, and is a potentially fatal CNS infec-
tion [94]. Kim et al. [95] have recently found that
CNS innate immunity and infection
group B Streptococcus infection of brain endothelial
Because of its low rates of neurogenesis, the CNS was cells induces upregulation of Snail1, a global repressor
long considered an ‘immune-privileged’ organ that of TJ gene expression. Snail1-mediated decreases in
required a BBB to prevent potentially damaging levels of claudin-5, occludin and ZO-1 in turn allow
immune cell entry and inflammation. However, it is for bacterial passage through the dysregulated TJ com-
increasingly being recognized that normal CNS physi- plexes [95]. Viral neuro-invasion has also been linked
ology involves continuous immune surveillance of to BBB disruption via TJ modulation. The West Nile
pathogen invasion and injury [9]. The BBB is integral virus (WNV) is a mosquito-borne flavivirus that can
to this process as the mechanisms of peripheral leuko- cause encephalitis (neuroinflammation), neuronal dys-
cyte contact with brain endothelial cells and migration function and death. Although the mechanisms of
across the BBB (also known as transendothelial leuko- WNV entry into the CNS are unclear, WNV-induced
cyte migration or TEM) become elucidated. In loss of major TJ proteins and increased matrix metal-
response to danger signals (such as tumour necrosis loproteinase production at the BBB have been
factor-a and interleukin-1b cytokines) on the ablumi- reported in WNV-infected mice [96]. A role for para-
nal or luminal side, brain endothelial cells become acti- cellular TJ disruption in WNV infiltration is also sup-
vated to release proinflammatory molecules and ported by the recent finding that interferon k
upregulate cell adhesion molecules like ICAM-1, treatment of brain endothelial cells stabilizes claudin-5
VCAM-1 and E-selectin [88]. Contact between periph- and ZO-1 localization after WNV infection [97].
eral leukocytes and brain endothelial cells is then Administration of interferon k in WNV-infected mice
mediated by interactions between cell adhesion mole- also reduced viral movement across the BBB [97].
cules and integrins like lymphocyte function-associated
antigen 1 on leukocytes [88]. Subsequent TEM is
Novel insights into BBB physiology
believed to occur paracellularly via reorganization of
the actin cytoskeleton and TJ remodelling, although Pathological changes in BBB function have now been
the precise mechanism is still unclear [89]. Rapid implicated in a wide range of CNS disorders [4], how-
remodelling of TJ complexes during TEM has recently ever less is known about the influence of common bio-
been reported using 3D confocal imaging of mono- logical processes on BBB development and integrity.
cytes during the TEM process [90]. In the space of Some intriguing work is pointing to the role of diverse
10 min, the addition of monocytes is followed by dis- phenomena such as sleep–wake behaviour and gut
continuous claudin-5 staining, specifically in areas of microbiota in establishing and maintaining a homeo-
monocyte TEM. These gaps are then rapidly resealed static BBB. We are also learning how the normal ageing
following leukocyte migration across the TJ [90]. process can impact BBB permeability and potentially
Intriguingly, barrier function was found to be pre- trigger disease-associated changes in cognitive function.

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4073


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

Sleep–wake cycle Ageing


Sleep is a universal and essential state among animals One of the strongest environmental risk factors for a
that is characterized by decreased sensory responsive- host of neurodegenerative conditions is age. BBB dys-
ness and reduced muscle tone [98]. The functions of function and vascular impairments have also been
sleep are still unclear although sleep likely assists in identified in the pathophysiology of many of these dis-
tissue repair, consolidation of memories and immune eases including AD and amyotrophic lateral sclerosis,
system function [98]. Sleep deprivation can lead to but whether these vascular changes are a cause or con-
impairments in neurocognitive function and increase sequence of neurodegeneration remains a question of
the risk of developing conditions like Type 2 diabetes active debate [105,106]. Some post-mortem histopatho-
[99]. Two recent studies have also found that sleep logical studies have shown that in the absence of
deprivation, in the form of chronic sleep restriction comorbidities, BBB integrity in humans declines with
[100] or selective rapid eye movement sleep deprivation age as evidenced by IgG extravasation, TJ protein
[101], can increase BBB permeability. Chronic sleep changes and cerebral microbleeds [107,108]. Further-
restriction, for example, was associated with reduced more, a meta-analysis of 31 BBB permeability studies
GLUT-1 and TJ protein expression at the BBB, (8 used post-mortem imaging, 21 of 23 others used
decreased brain glucose uptake and increased paracel- CSF/plasma albumin ratio) of normal ageing found
lular permeability to sodium fluorescein and biotin tra- that BBB permeability increased with age [109],
cer molecules [100]. In both studies, sleep recovery although these imaging and biochemical approaches
restored BBB structure and function including a return have limitations including tissue processing and CSF
to baseline TJ protein levels [100,101]. Related work turnover rates [110]. New work by Montagne and col-
has also elucidated the role of sleep in regulating the leagues [111] has used a high-resolution method of
exchange of solutes like Ab between the cerebrospinal dynamic contrast-enhanced MRI to assess BBB perme-
fluid (CSF) and interstitial fluid along paravenous ability in the living human brain. In the CA1 and den-
drainage pathways (the so-called ‘glymphatic’ system) tate gyrus regions of the hippocampus, they found an
[102]. These combined findings suggest that sleep regu- age-dependent BBB breakdown in cognitively normal
lation of cerebrovascular clearance pathways is vital to individuals (from 23 to 91 years of age) based on the
maintaining brain homeostasis. blood-to-brain transfer constant (Ktrans) of gadolinium
contrast agent [111]. No obvious age-related changes
in BBB permeability were seen in cortical and subcor-
Gut microbiota
tical regions or in white matter. BBB permeability in
The collection of microorganisms in the human gas- the hippocampus was further increased in individuals
trointestinal tract is known as the gut microbiota. with mild cognitive impairment (a disorder that can
There is accumulating evidence that gut microbiota precede AD) leading the authors to suggest that age-
can communicate with the CNS to alter neural func- ing-related BBB breakdown may contribute to demen-
tion and behaviour via changes in the autonomic ner- tia onset [111]. CSF analyses also showed that CSF
vous system, microbial metabolites and/or systemic levels of sPDGFRb, a potential biomarker of pericyte
inflammatory cytokines levels [103]. Braniste and col- injury, increased with age [111]. Because pericyte loss
leagues [104] have recently shown that a lack of gut and dysfunction in murine models have been associ-
microbiota is associated with reduced levels of claudin- ated with BBB breakdown [23,25,35], increased BBB
5 and occludin and increased BBB permeability. permeability in the ageing human hippocampus may
Interestingly, this phenotype was sustained from devel- be due to pericyte damage. For future research, this
opment into adulthood indicating that BBB function novel dynamic contrast-enhanced MRI method pro-
may depend on constant communication with the gut vides an exciting platform to investigate in ‘real time’
microbiota throughout life. The authors also found how genetics, diet and medicines can affect BBB integ-
that exposure of germ-free mice to Clostridium tyrobu- rity in living humans.
tyricum, a bacterial strain that produces butyrate, or
administration of sodium butyrate alone upregulates
Conclusions
TJ protein expression and restores BBB tightness
[104]. Although the mechanism is still largely unclear, Whereas transplantation and histological studies domi-
isolating the factor(s) responsible for gut microbiota nated early BBB research, our knowledge of how
regulation of BBB integrity may provide novel means NVU components interact at the molecular level to
of restoring a leaky BBB in disease. signal BBB development and maintenance has

4074 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

advanced over the last decade. It is clear that early formation and function of the blood-brain barrier.
and sustained communication in the NVU via a range Nature 509, 507–511.
of secreted morphogens and cell receptors gives rise to 8 Luissint AC, Artus C, Glacial F, Ganeshamoorthy K
and maintains the unique barrier properties of the & Couraud PO (2012) Tight junctions at the blood
BBB. Furthermore, dysfunction of any one NVU brain barrier: physiological architecture and
member can ultimately lead to dysregulation of tran- disease-associated dysregulation. Fluids Barriers CNS
scellular and paracellular pathways across brain 9, 9–23.
endothelial cells and contribute to pathological BBB 9 Lampron A, Elali A & Rivest S (2013) Innate
immunity in the CNS: redefining the relationship
breakdown. Much work to date has focused on the
between the CNS and its environment. Neuron 78,
role of astrocytes and pericytes in BBB regulation.
214–232.
Elucidating how other cell types, in particular neurons
10 Stewart PA & Wiley MJ (1981) Developing nervous
and microglia, interact with brain endothelial cells
tissue induces formation of blood-brain barrier
may offer further clues to the complete recipe for a
characteristics in invading endothelial cells: a study
fully functional BBB and to repairing the damaged using quail-chick transplantation chimeras. Dev Biol
barrier. Conversely, new methods of circumventing the 84, 183–192.
BBB to deliver small or large molecule drugs into the 11 Rodrıguez-Arellano JJ, Parpura V, Zorec R &
brain could take advantage of the dynamic nature of Verkhratsky A (2015) Astrocytes in physiological
TJ complexes. To conclude, as we increase our under- aging and Alzheimer’s disease. Neuroscience. doi:
standing of BBB involvement in brain homeostasis 10.1016/j.neuroscience.2015.01.007 (in press).
and disease, it highlights the importance of considering 12 Abbott NJ, R€ onnb€ack L & Hansson E (2006)
BBB biology in all basic and translational neuroscience Astrocyte-endothelial interactions at the blood-brain
research. barrier. Nat Rev Neurosci 7, 41–53.
13 Zlokovic BV (2008) The blood-brain barrier in health
and chronic neurodegenerative disorders. Neuron 57,
Author Contributions 178–195.
James Keaney and Matthew Campbell wrote the 14 Tait MJ, Saadoun S, Bell BA & Papadopoulos MC
review. (2008) Water movements in the brain: role of
aquaporins. Trends Neurosci 31, 37–43.
15 Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I,
References Fabre PJ, Terouz S, Sabbagh M, Wosik K,
1 Abbott NJ, Patabendige AAK, Dolman DEM, Yusof Bourbonniere L, Bernard M et al. (2011) The
SR & Begley DJ (2010) Structure and function of the hedgehog pathway promotes blood-brain barrier
blood-brain barrier. Neurobiol Dis 37, 13–25. integrity and CNS immune quiescence. Science 334,
2 Saunders NR, Liddlelow SA & Dziegielewska KM 1727–1731.
(2012) Barrier mechanisms in the developing brain. 16 Wosik K, Cayrol R, Dodelet-Devillers A, Berthelet F,
Front Pharmacol 3, 1–18. Bernard M, Moumdjian R, Bouthillier A, Reudelhuber
3 Engelhardt B & Liebner S (2014) Novel insights into TL & Prat A (2007) Angiotensin II controls occludin
the development and maintenance of the blood-brain function and is required for blood-brain barrier
barrier. Cell Tissue Res 355, 687–699. maintenance: relevance to multiple sclerosis. J Neurosci
4 Obermeier B, Daneman R & Ransohoff R (2013) 27, 9032–9042.
Development, maintenance and disruption of the 17 Bell RD, Winkler EA, Singh I, Sagare AP, Deane R,
blood-brain barrier. Nat Med 19, 1584–1596. Wu Z, Holtzman DM, Betsholtz C, Armulik A,
5 Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell Sallstrom J et al. (2012) Apolipoprotein E controls
RD, Perlmutter D, Sengillo JD, Hillman S, Kong P, cerebrovascular integrity via cyclophilin A. Nature 485,
Nelson AR et al. (2015) GLUT1 reductions exacerbate 512–516.
Alzheimer’s disease vasculo-neuronal dysfunction and 18 Nishitsuji K, Hosono T, Nakamura T, Bu G &
degeneration. Nat Neurosci 18, 521–530. Michikawa M (2011) Apolipoprotein E regulates the
6 Nguyen LN, Ma D, Shui G, Wong P, Cazenave-Gassiot integrity of tight junctions in an isoform-dependent
A, Zhang X, Wenk MR, Goh EL & Silver DL (2014) manner in an in vitro blood-brain barrier model. J
Mfsd2a is a transporter for the essential omega-3 fatty Biol Chem 286, 17536–17542.
acid docosahexaenoic acid. Nature 509, 503–506. 19 Janzer RC & Raff MC (1987) Astrocytes induce
7 Ben-Zvi A, Lacoste B, Kur E, Andreone BJ, Mayshar blood-brain barrier properties in endothelial cells.
Y, Yan H & Gu C (2014) Mfsd2a is critical for the Nature 325, 253–257.

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4075


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

20 Holash JA, Noden DM & Stewart PA (1993) Re- promoting ligand-specific canonical wnt signaling. Dev
evaluating the role of astrocytes in blood-brain barrier Cell 31, 248–256.
induction. Dev Dyn 197, 14–25. 33 Lindahl P, Johansson BR, Leveen P & Betsholtz C
21 Dehouck MP, Meresse S, Delorme P, Fruchart JC & (1997) Pericyte loss and microaneurysm formation in
Cecchelli R (1990) An easier, reproducible, and mass- PDGF-B–deficient mice. Science 277, 242–245.
production method to study the blood–brain barrier 34 Li F, Lan Y, Wang Y, Wang J, Yang G, Meng F,
in vitro. J Neurochem 54, 1798–1801. Han H, Meng A, Wang Y & Yang X (2011)
22 Hayashi Y, Nomura M, Yamagishi S, Harada S, Endothelial Smad4 maintains cerebrovascular integrity
Yamashita J & Yamamoto H (1997) Induction of by activating N-cadherin through cooperation with
various blood-brain barrier properties in non-neural Notch. Dev Cell 20, 291–302.
endothelial cells by close apposition to co-cultured 35 Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B,
astrocytes. Glia 19, 13–26. Deane R & Zlokovic B (2010) Pericytes control key
23 Daneman R, Zhou L, Kebede AA & Barres BA (2010) neurovascular functions and neuronal phenotype in the
Pericytes are required for blood-brain barrier integrity adult brain and during brain aging. Neuron 68, 409–427.
during embryogenesis. Nature 468, 562–566. 36 Armulik A, Genove G, Mae M, Nisancioglu MH,
24 Mizee MR, Wooldrik D, Lakeman KA, van het Hof Wallgard E, Niaudet C, He L, Norlin J, Lindblom P,
B, Drexhage JA, Geerts D, Bugiani M, Aronica E, Strittmatter K et al. (2010) Pericytes regulate the
Mebius RE, Prat A et al. (2013) Retinoic acid induces blood-brain barrier. Nature 468, 557–561.
blood-brain barrier development. J Neurosci 33, 1660– 37 Sagare AP, Bell RD, Zhao Z, Ma Q, Winkler EA,
1671. Ramanathan A & Zlokovic BV (2013) Pericyte loss
25 Winkler EA, Bell RD & Zlokovic BV (2011) Central influences Alzheimer-like neurodegeneration in mice.
nervous system pericytes in health and disease. Nat Nat Commun 4, 2932.
Neurosci 14, 1398–1405. 38 Ginhoux F, Greter M, Leboeuf M, Nandi S, See P &
26 Hall CN, Reynell C, Gesslein B, Hamilton NB, Gokhan S (2010) Fate mapping analysis reveals that
Mishra A, Sutherland BA, O’Farrell FM, Buchan adult microglia derive from primitive macrophages.
AM, Lauritzen M & Atwell D (2014) Capillary Science 330, 841–845.
pericytes regulate cerebral blood flow in health and 39 Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q,
disease. Nature 508, 55–60. Prykhozhij S, Peri F, Wilson SW & Ruhrberg C (2010)
27 Liebner S, Corada M, Bangsow T, Babbage J, Taddei Tissue macrophages act as cellular chaperones for
A, Czupolla CJ, Reis M, Felici A, Wolburg H, vascular anastomosis downstream of VEGF-mediated
Fruttiger M et al. (2008) Wnt/b-catenin signaling endothelial tip cell induction. Blood 116, 829–840.
controls development of the blood-brain barrier. J Cell 40 da Fonseca AC, Matias D, Garcia C, Amaral R,
Biol 183, 409–417. Geraldo LH, Freitas C & Lima FR (2014) The impact
28 Stenman J, Rajagopal J, Carroll TJ, Ishibashi M, of microglial activation on blood-brain barrier in brain
McMahon J & McMahon AP (2008) Canonical Wnt diseases. Front Cell Neurosci 8, 362.
signaling regulates organ-specific assembly and 41 Aguzzi A, Barres BA & Bennett ML (2013) Microglia:
differentiation of CNS vasculature. Science 322, 1247– scapegoat, saboteur, or something else? Science 339,
1250. 156–161.
29 Daneman R, Agalliu D, Zhou L, Kuhnert F, Kuo CJ 42 Fischer MT, Sharma R, Lim JL, Haider L, Frischer
& Barres BA (2009) Wnt/b-catenin is required for JM, Drexhage J, Mahad D, Bradl M, van Horssen J &
CNS, but not non-CNS, angiogenesis. Proc Natl Acad Lassmann H (2012) NADPH oxidase expression in
Sci USA 106, 641–646. active multiple sclerosis lesions in relation to oxidative
30 Tam SJ, Richmond DL, Kaminker JS, Modrusen Z, tissue damage and mitochondrial injury. Brain 135,
Martin-McNulty B, Cao TC, Weimer RW, Carano 886–899.
RAD, van Bruggen N & Watts R (2012) Death 43 Rochfort KD, Collins LE, Murphy RP & Cummins
receptors DR6 and TROY regulate brain vascular PM (2014) Downregulation of blood-brain barrier
development. Dev Cell 22, 403–417. phenotype by proinflammatory cytokines involves
31 Cullen M, Elzarrad MK, Seaman S, Zudaire E, NADPH oxidase-dependent ROS generation:
Stevens J, Yang MY, Li X, Chaudhary A, Xu L, consequences for interendothelial adherens and tight
Hilton MB et al. (2011) GPR124, an orphan G junctions. PLoS ONE 9, e101815.
protein-coupled receptor, is required for CNS-specific 44 Yang YM, Shang DS, Zhao WD, Fang WG & Chen
vascularization and establishment of the blood-brain YH (2013) Microglial TNF-a-dependent elevation of
barrier. Proc Natl Acad Sci USA 108, 5759–5764. MHC class I expression on brain endothelium induced
32 Zhou Y & Nathans J (2014) Gpr124 controls CNS by amyloid-beta promotes T cell transendothelial
angiogenesis and blood-brain barrier integrity by migration. Neurochem Res 38, 2295–2304.

4076 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

45 Yin X, Wright J, Wall T & Grammas P (2010) 58 Roberts KF, Elbert DL, Kasten TP, Patterson BW,
Brain endothelial cells synthesize neurotoxic Sigurdson WC, Connors RE, Ovod V, Munsell LY,
thrombin in Alzheimer’s disease. Am J Pathol 176, Mawuenyega KG, Miller-Thomas MM et al. (2014)
1600–1606. Amyloid-b efflux from the central nervous system into
46 Huang CF, Li G, Ma R, Sun SG & Chen JG (2008) the plasma. Ann Neurol 76, 837–844.
Thrombin-induced microglial activation contributes to 59 Cirrito JR, Deane R, Fagan AM, Spinner ML,
the degeneration of nigral dopaminergic neurons Parsadanian M, Finn MB, Jiang H, Prior JL, Sagare
in vivo. Neurosci Bull 24, 66–72. A, Bales KR et al. (2005) P-glycoprotein deficiency at
47 Hickey WF & Kimura H (1988) Perivascular the blood-brain barrier increases amyloid-beta
microglial cells of the CNS are bone marrow-derived deposition in an Alzheimer disease mouse model. J
and present antigen in vivo. Science 239, 290–292. Clin Invest 115, 3285–3290.
48 Yao Y, Chen ZL, Norris EH & Strickland S (2014) 60 Kuhnke D, Jedlitschky G, Grube M, Krohn M, Jucker
Astrocytic laminin regulates pericyte differentiation M, Mosyagin I, Cascorbi I, Walker LC, Kroemer HK,
and maintains blood brain barrier integrity. Nat Warzok RW et al. (2007) MDR1-P-Glycoprotein
Commun 5, 3413. (ABCB1) mediates transport of Alzheimer’s amyloid-
49 Pardridge WM (2009) Alzheimer’s disease drug beta peptides-implications for the mechanisms of
development and the problem of the blood-brain Abeta clearance at the blood-brain barrier. Brain
barrier. Alzheimer’s Dement 5, 427–432. Pathol 17, 347–353.
50 Harik SI, Kalaria RN, Andersson L, Lundahl P & 61 Deane R, Du Yan S, Submamaryan RK, LaRue B,
Perry G (1990) Immunocytochemical localization of Jovanovic S, Hogg E, Welch D, Manness L, Lin C,
the erythroid glucose transporter: abundance in tissues Yu J et al. (2003) RAGE mediates amyloid-beta
with barrier functions. J Neurosci 10, 3862–3872. peptide transport across the blood-brain barrier and
51 Rotstein M, Engelstad K, Yang H, Wang D, Levy B, accumulation in brain. Nat Med 9, 907–913.
Chung WK & De Vivo DC (2010) Glut1 deficiency: 62 Kanekiyo T, Cirrito JR, Liu CC, Shinohara M, Li J,
inheritance pattern determined by haploinsufficiency. Schuler DR, Shinohara M, Holtzman DM & Bu G
Ann Neurol 68, 955–958. (2013) Neuronal clearance of amyloid-beta by
52 Mooradian AD, Chung HC & Shah GN (1997) endocytic receptor LRP1. J Neurosci 33, 19276–19283.
GLUT1 expression in the cerebra of patients with 63 Kanekiyo T, Liu CC, Shinohara M, Li J & Bu G
Alzheimer’s disease. Neurobiol Aging 18, 469–474. (2012) LRP1 in brain vascular smooth muscle cells
53 Landau SM, Harvey D, Madison CM, Koeppe RA, mediates local clearance of Alzheimer’s amyloid-beta.
Reiman EM, Foster NL, Weiner MW, Jagust WJ & J Neurosci 32, 16458–16465.
Alzheimer’s Disease Neuroimaging Initiative (2011) 64 Deane R, Wu Z, Sagare A, Davis J, Du Yan S,
Associations between cognitive, functional, and FDG- Hamm K, Xu F, Parisi M, LaRue B, Hu HW et al.
PET measures of decline in AD and MCI. Neurobiol (2004) LRP/amyloid beta-peptide interaction mediates
Aging 32, 1207–1218. differential brain efflux of Abeta isoforms. Neuron 43,
54 Hunt A, Sch€ onknecht P, Henze M, Seidl U, 333–344.
Haberkorn U & Schr€ oder J (2007) Reduced cerebral 65 Bell RD, Deane R, Chow N, Long X, Sagare A, Singh
glucose metabolism in patients at risk for Alzheimer’s I, Streb JW, Guo H, Rubio A, Van Nostrand W et al.
disease. Psychiatry Res 155, 147–154. (2009) SRF and myocardin regulate LRP-mediated
55 Herholz K (2010) Cerebral glucose metabolism in amyloid-beta clearance in brain vascular cells. Nat Cell
preclinical and prodromal Alzheimer’s disease. Expert Biol 11, 143–153.
Rev Neurother 10, 1667–1673. 66 Zhao Z, Sagare AP, Ma Q, Halliday MR, Kong P,
56 Alakbarzade V, Hameed A, Quek DQ, Chioza BA, Kisler K, Winkler EA, Ramanathan A, Kanekiyo T,
Baple EL, Cazenave-Gassiot A, Nguyen LN, Wenk Bu G et al. (2015) Central role for PICALM in
MR, Ahmad AQ, Sreekantan-Nair A et al. (2015) A amyloid-b blood-brain barrier transcytosis and
partially inactivating mutation in the sodium- clearance. Nat Neurosci 18, 978–987.
dependent lysophosphatidylcholine transporter 67 Harold D, Abraham R, Hollingworth P, Sims R,
MFSD2A causes a non-lethal microcephaly syndrome. Gerrish A, Hamshere ML, Pahwa JS, Moskvina V,
Nat Genet. doi:10.1038/ng.3313. Dowzell K, Williams A et al. (2009) Genome-wide
57 Guemez-Gamboa A, Nguyen LN, Yang H, Zaki MS, association study identifies variants at CLU and
Kara M, Ben-Omran T, Akizu N, Rosti RO, Rosti B, PICALM associated with Alzheimer’s disease. Nat
Scott E et al. (2015) Inactivating mutations in Genet 41, 1088–1093.
MFSD2A, required for omega-3 fatty acid transport in 68 Bazzoni G & Dejana E (2004) Endothelial cell-to-cell
brain, cause a lethal microcephaly syndrome. Nat junctions: molecular organization and role in vascular
Genet. doi:10.1038/ng.3311. homeostasis. Physiol Rev 84, 869–901.

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4077


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
The dynamic blood–brain barrier J. Keaney and M. Campbell

69 Hawkins BT & Davis TP (2005) The blood-brain 81 Miyazaki K, Ohta Y, Nagai M, Morimoto N, Kurata
barrier/neurovascular unit in health and disease. T, Takehisa Y, Ikeda Y, Matsuura T & Abe K (2011)
Pharmacol Rev 57, 173–185. Disruption of neurovascular unit prior to motor
70 Tietz S & Engelhardt B (2015) Brain barriers: crosstalk neuron degeneration in amyotrophic lateral sclerosis. J
between complex tight junctions and adherens Neurosci Res 89, 718–728.
junctions. J Cell Biol 4, 493–506. 82 Hartz AMS, Bauer B, Soldner LB, Wolf A, Boy S,
71 Haseloff RF, Dithmer S, Winkler L, Wolburg H & Backhaus R, Mihaljevic I, Bogdahn U, Klunemann
Blasig IE (2015) Transmembrane proteins of the tight HH, Schuierer G et al. (2011) Amyloid-beta
junctions at the blood-brain barrier: Structural and contributes to blood-brain barrier leakage in
functional aspects. Semin Cell Dev Biol 38, 16–25. transgenic human amyloid precursor protein mice and
72 Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal in humans with cerebral amyloid angiopathy. Stroke
A & Barres BA (2010) The mouse blood-brain barrier 43, 514–523.
transcriptome: a new resource for understanding the 83 Khatri R, McKinney AM, Swenson B & Janardhan V
development and function of brain endothelial cells. (2012) Blood-brain barrier, reperfusion injury, and
PLoS ONE 5, 1–16. hemorrhagic transformation in acute ischemic stroke.
73 Fanning AS, Jameson BJ, Jesaitis LA & Anderson JM Neurology 79, S52–S57.
(1998) The tight junction protein ZO-1 establishes a 84 Sandoval KE & Witt KA (2008) Blood-brain barrier
link between the transmembrane protein occludin and tight junction permeability and ischemic stroke.
the actin cytoskeleton. J Biol Chem 273, 29745–29753. Neurobiol Dis 32, 200–219.
74 Iwamoto N, Higashi T & Furuse M (2014) 85 Knowland D, Arac A, Sekiguchi KJ, Hsu M, Lutz SE,
Localization of angulin-1/LSR and tricellulin at Perrino J, Steinberg GK, Barres BA, Nimmerjahn A &
tricellular contacts of brain and retinal endothelial cells Agalliu D (2014) Stepwise recruitment of transcellular
in vivo. Cell Struct Funct 39, 1–8. and paracellular pathways underlies blood-brain
75 Sohet F, Lin C, Munji RN, Lee SY, Ruderisch N, barrier breakdown in stroke. Neuron 82, 603–617.
Soung A, Arnold TD, Derugin N, Vexler ZS, Yen FT 86 Nag S, Venugopalan R & Stewart DJ (2007) Increased
et al. (2015) LSR/angulin-1 is a tricellular tight caveolin-1 expression precedes decreased expression of
junction protein involved in blood-brain barrier occludin and claudin-5 during blood-brain barrier
formation. J Cell Biol 208, 703–711. breakdown. Acta Neuropathol 114, 459–469.
76 Tornavaca O, Chia M, Dufton N, Almagro LO, 87 Campbell M, Hanrahan F, Gobbo OL, Kelly ME,
Conway DE, Randi AM, Schwartz MA, Matter K & Kiang AS, Humphries MM, Nguyen ATH, Ozaki E,
Balda MS (2015) ZO-1 controls endothelial adherens Keaney J, Blau CW et al. (2012) Targeted suppression
junctions, cell-cell tension, angiogenesis, and barrier of claudin-5 decreases cerebral oedema and improves
formation. J Cell Biol 208, 821–838. cognitive outcome following traumatic brain injury.
77 Taddei A, Giampietro C, Conti A, Orsenigo F, Nat Commun 3, Article 849.
Breviario F, Pirazzoli V, Potente M, Daly C, 88 Greenwood J, Wang Y & Calder VL (1995)
Dimmeler S & Dejana E (2008) Endothelial adherens Lymphocyte adhesion and transendothelial migration
junctions control tight junctions by VE-cadherin- in the central nervous system: the role of LFA-1,
mediated upregulation of claudin-5. Nat Cell Biol 10, ICAM-1, VLA-4 and VCAM-1. Immunology 86, 408–
923–934. 415.
78 Walsh TG, Murphy RP, Fitzpatrick P, Rochfort KD, 89 Sagar D, Lamontagne A, Foss CA, Khan ZK, Pomper
Guinan AF, Murphy A & Cummins PM (2011) MG & Jain P (2012) Dendritic cell CNS recruitment
Stabilization of brain microvascular endothelial barrier correlates with disease severity in EAE via CCL2
function by shear stress involves VE-cadherin signaling chemotaxis at the blood-brain barrier through
leading to modulation of pTyr-occludin levels. J Cell paracellular transmigration and ERK activation. J
Physiol 226, 3053–3063. Neuroinflammation 9, 245.
79 Alvarez JI, Saint-Laurent O, Godschalk A, Terouz S, 90 Winger RC, Koblinski JE, Kanda T, Ransohoff RM
Briels C, Larouche S, Bourbonniere L, Larochelle C & & Muller WA (2014) Rapid remodeling of tight
Prat A (2015) Focal disturbances in the blood-brain junctions during paracellular diapedesis in a human
barrier are associated with formation of model of the blood-brain barrier. J Immunol 193,
neuroinflammatory lesions. Neurobiol Dis 74, 14–24. 2427–2437.
80 Zhong Z, Deane R, Ali Z, Parisi M, Shapovalov Y, 91 Larochelle C, Alvarez JI & Prat A (2011) How do
O’Banion MK, Stojanovic K, Sagare A, Boillee S, immune cells overcome the blood-brain barrier in
Cleveland DW et al. (2008) ALS-causing SOD1 multiple sclerosis? FEBS Lett 585, 3770–3780.
mutants generate vascular changes prior to motor 92 Simard AR, Soulet D, Gowing G, Julien JP & Rivest
neuron degeneration. Nat Neurosci 11, 420–422. S (2006) Bone marrow-derived microglia play a critical

4078 FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS


17424658, 2015, 21, Downloaded from https://febs.onlinelibrary.wiley.com/doi/10.1111/febs.13412 by CAPES, Wiley Online Library on [06/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J. Keaney and M. Campbell The dynamic blood–brain barrier

role in restricting senile plaque formation in 102 Xie L, Kang H, Xu Q, Chen MJ, Liao Y,
Alzheimer’s disease. Neuron 49, 489–502. Thiyagarajan M, O’Donnell J, Christensen DJ,
93 Ziv Y, Ron N, Butovsky O, Landa G, Sudai E, Nicholson C, Iliff JJ et al. (2013) Sleep drives
Greenberg N, Cohen H, Kipnis J & Schwartz M metabolic clearance from the adult brain. Science 342,
(2006) Immune cells contribute to the maintenance of 373–377.
neurogenesis and spatial learning abilities in 103 Cryan JF & Dinan TG (2012) Mind-altering
adulthood. Nat Neurosci 9, 268–275. microorganisms: the impact of the gut microbiota on
94 Saez-Llorens X & McCracken GH Jr (2003) Bacterial brain and behaviour. Nat Rev Neurosci 13, 701–712.
meningitis in children. Lancet 361, 2139–2148. 104 Braniste V, Al-Asmakh M, Kowal C, Anuar F,
95 Kim BJ, Hancock BM, Bermudez A, Cid ND, Reyes Abbaspour A, T oth M, Korecka A, Bakocevic N, Ng
E, vanSorge NM, Lauth X, Smurthwaite CA, Hilton LG, Kundu P et al. (2014) The gut microbiota
BJ, Stotland A et al. (2015) Bacterial induction of influences blood-brain barrier permeability in mice. Sci
Snail1 contributes to blood-brain barrier disruption. J Transl Med 6, 263ra158.
Clin Invest 125, 2473–2483. 105 Zlokovic BV (2011) Neurovascular pathways to
96 Roe K, Kumar M, Lum S, Orillo B, Nerurkar VR & neurodegeneration in Alzheimer’s disease and other
Verma S (2012) West Nile virus-induced disruption of disorders. Nat Rev Neurosci 12, 723–738.
the blood-brain barrier in mice is characterized by the 106 Erickson MA & Banks WA (2013) Blood-brain barrier
degradation of the junctional complex proteins and dysfunction as a cause and consequence of
increase in multiple matrix metalloproteinases. J Gen Alzheimer’s disease. J Cereb Blood Flow Metab 33,
Virol 93, 1193–1203. 1500–1513.
97 Lazear HM, Daniels BP, Pinto AK, Huang AC, Vick 107 Bake S, Friedman JA & Sohrabji F (2009)
SC, Doyle SE, Gale MJr , Klein RS & Diamond MS Reproductive age-related changes in the blood brain
(2015) Interferon-k restricts West Nile virus barrier: expression of IgG and tight junction proteins.
neuroinvasion by tightening the blood-brain barrier. Microvasc Res 78, 413–424.
Sci Transl Med 7, 284ra59. 108 De Reuck JL (2012) Histopathological stainings and
98 Siegel JM (2005) Clues to the functions of mammalian definitions of vascular disruptions in the elderly brain.
sleep. Nature 437, 1264–1271. Exp Gerontol 47, 834–837.
99 Kawakami N, Takatsuka N & Shimizu H (2004) Sleep 109 Farrall AJ & Wardlaw JM (2009) Blood-brain barrier:
disturbance and onset of type 2 diabetes. Diabetes ageing and microvascular disease–systematic review
Care 27, 282–283. and meta-analysis. Neurobiol Aging 30, 337–352.
100 G omez-Gonzalez B, Hurtado-Alvarado G, Esqueda- 110 Chen RL (2011) Is it appropriate to use albumin CSF/
Le on E, Santana-Miranda R, Rojas-Zamorano JA  & plasma ratio to assess blood brain barrier
Velazquez-Moctezuma J (2013) REM sleep loss and permeability? Neurobiol Aging 32, 1338–1339.
recovery regulates blood-brain barrier function. Curr 111 Montagne A, Barnes SR, Sweeney MD, Halliday MR,
Neurovasc Res 10, 197–207. Sagare AP, Zhao Z, Toga AW, Jacobs RE, Liu CY,
101 He J, Hsuchou H, He Y, Kastin AJ, Wang Y & Pan Amezcua L et al. (2015) Blood-brain barrier
W (2015) Sleep restriction impairs blood-brain barrier breakdown in the aging human hippocampus. Neuron
function. J Neurosci 34, 14697–14706. 85, 296–302.

FEBS Journal 282 (2015) 4067–4079 ª 2015 FEBS 4079

You might also like