SlideShare a Scribd company logo
1 of 9
Download to read offline
Microreview
How can microbial interactions with the blood–brain
barrier modulate astroglial and neuronal function?
Dennis J. Grab,1
Srabasti J. Chakravorty,2
Henri van der Heyde3
and Monique F. Stins4
*
1
Johns Hopkins SOM, Department Pathology, Division
Medical Microbiology, 720 Rutland Ave, Ross 624,
Baltimore, MD 21205, USA.
2
Institute for Science and Technology in Medicine, Keele
University, Keele, Staffordshire, UK.
3
La Jolla Infectious Disease Institute, Bunker Hill Rd, La
Jolla, CA, USA.
4
Johns Hopkins SOM, Department Neurology,
NeuroSurgery and Psychiatry, Division
NeuroImmunology, 600 N Wolfe street, Meyer 5-134
(mail 6-113), Baltimore, MD 21281, USA.
Summary
The vascular endothelium of the blood–brain
barrier (BBB) is regarded as a part of the neurovas-
cular unit (NVU). This emerging NVU concept
emphasizes the need for homeostatic signalling
among the neuronal, glial and vascular endothelial
cellular compartments in maintaining normal brain
function. Conversely, dysfunction in any compo-
nent of the NVU affects another, thus contributing
to disease. Brain endothelial activation and dys-
function is observed in various neurological dis-
eases, such as (ischemic) stroke, seizure, brain
inflammation and infectious diseases and likely
contributes to or exacerbates neurological condi-
tions. The role and impact of brain endothelial
factors on astroglial and neuronal activation is
unclear. Similarly, it is not clear which stages of
BBB endothelial activation can be considered
beneficial versus detrimental. Although the BBB
plays an important role in context of encephalopa-
thies caused by neurotropic microbes that must
first penetrate into the brain, a crucial role of the
BBB in contributing to neurological dysfunction
may be seen in cerebral malaria (CM), where the
Plasmodium parasite remains sequestered in the
brain vasculature, does not enter the brain paren-
chyma, and yet causes coma and seizures. In this
minireview some of the scenarios and factors that
may play a role in BBB as a relay station to modu-
late astroneuronal functioning are discussed.cmi_1661 1470..1478
The blood–brain barrier endothelium and the
neurovascular unit
The blood–brain barrier (BBB) endothelium forms a highly
selective barrier between the blood and the central
nervous system (CNS). Once regarded as ‘just’ a lining for
vessels to allow blood to flow and nutrients to pass into
the underlying tissues, it is now clear that endothelium in
different tissues is very heterogeneous in its expression of
surface receptors and responses to stimuli (Langenkamp
and Molema, 2009). Contrary to liver endothelium that
displays fenestrae to facilitate plasma exchange, brain
endothelium possesses tight junctions characteristic of its
barrier function. The unique tissue-specific endothelial
characteristics are induced through interactions with the
underlying tissues and in the CNS this includes the under-
lying extracellular matrix, astrocytes and pericytes (Abbott
et al., 2006; Kroll et al., 2009; Bonkowski et al., 2011;
Kamouchi et al., 2011). In addition, brain-trophic BBB-
derived factors can promote differentiation of neuropre-
cursor cells (Guo et al., 2008; Chintawar et al., 2009).
Indeed, the BBB endothelium (BBB-EC) is now regarded
an integral component of the neurovascular unit (NVU),
an emerging concept that underlines the importance of
the homeostatic interactions between the brain’s cellular
components – astroglia, neurons and BBB-EC – for
optimal CNS functioning (Abbott et al., 2006; Persidsky
et al., 2006; Lok et al., 2007; Bonkowski et al., 2011).
Conversely, deregulation in any of the NVU components,
will alter the brains homeostasis leading to a multitude of
neurodysfunctions and BBB activation as has been
observed in multiple sclerosis, Alzheimers disease,
stroke, certain depression disorders and in microbial
infections, among others (Zlokovic, 2008; Reale et al.,
2009; Chung et al., 2010; Holman et al., 2011). The
Received 21 June, 2011; revised 2 August, 2011; accepted 3 August,
2011. *For correspondence. E-mail mstins@jhmi.edu; Tel. (+1)
443 287 8027
Cellular Microbiology (2011) 13(10), 1470–1478 doi:10.1111/j.1462-5822.2011.01661.x
First published online 6 September 2011
© 2011 Blackwell Publishing Ltd
cellular microbiology
contribution of several endothelial-activating components,
including microbial involvement, has been suggested to
initiate certain brain diseases (Miklossy, 2008; Zhang
et al., 2009; Humpel, 2011; Ochoa-Reparaz et al., 2011).
Circulating gut-derived lipopolysaccharides (LPS), which
are increased in, e.g. HIV-1 infection, alcohol abuse and
depression disorders, can activate brain endothelium to
trigger astroglial activation signals and/or modulate neu-
rological functioning that lead to sickness-related behav-
ioural changes (Schafer et al., 2002; Maes et al., 2008).
Modulation of microglial function by peripheral inflamma-
tion also depends on the prior history and immune status
(Cunningham et al., 2009). Unfortunately, the role of
BBB-EC activation in the development of neurological
diseases is still an understudied area of research.
In vitro BBB endothelial models to study its
involvement in neurological disease
Single- or multi-cell in vitro models of the BBB have
proven useful tools to study BBB-EC and its functionality
in neurological diseases. For elucidation of specific BBB
human endothelium responses, cultures comprising of
only primary or immortalized human brain EC are pre-
ferred (Stins et al., 2001a; Weksler et al., 2005). However,
other models include non-human- and non-brain-
endothelial cell types such as human umbilical vein endot-
helial cells (Weiss et al., 1999; Eugenin et al., 2006),
ECV304/T4 cells (Tan et al., 2001; Neuhaus et al., 2011),
primary porcine brain endothelial cultures (Kroll et al.,
2009), immortalized rat cultures (Roux and Couraud,
2005) and bovine brain endothelial cells (Stins et al.,
1997). Today, a wide availability of diverse immortalized
cell lines have become attractive workhorses and have
greatly facilitated BBB research. However, because
immortalized lines may have changed their physiology
and do not always reflect the heterogeneity of endothelial
populations and responses, it is imperative to verify
results with primary brain endothelial cells that are now
commercially available. With these tools, research to
better understand the intimate interactions between
BBB-EC and other cellular components within NVU and
its contributions to neurological disease is now universally
possible.
To elicit more BBB-like characteristics, BBB-EC can be
cultured on brain-specific extracellular matrix, in combina-
tion multi-cell co-cultures, either with pericytes, astro-
cytes, C-6 glioma cells or in the presence of conditioned
media from these cells (Nakagawa et al., 2009; Hatherell
et al., 2011). The ‘modular’ system approach allows
studies on direct interactions between the different cell
types of the NVU, which may not be possible in vivo.
However, astrocytes are heterogeneous and can be ‘acti-
vated’ upon isolation (Zhang and Barres, 2010), either
positively or negatively impacting on the BBB-EC charac-
teristics when used in co-culture models. Physical factors,
such as vascular sheer stress and red blood cell (RBC)
interactions, can affect BBB characteristics such as
higher trans endothelial electrical resistance (TEER),
decreased permeability and altered expression of multi-
drug transporters (Tripathi et al., 2007; Cucullo et al.,
2011a,b). Complex hollow tube dynamic flow models
and two-dimensional flow models allow for studies on
interactions of (microbial infected-) immune cells and/or
RBCs under flow (Barth et al., 2009; Phiri et al., 2009)
with BBB-EC.
Microbes, BBB and neurological dysfunction; to
cross or not to cross?
Generally, high plasma concentrations of neurotropic
microbes (e.g. sepsis) are a prerequisite for CNS entry.
BBB crossing can occur via transcellular or paracellular
mechanisms, or when immune cells are ‘hijacked’ by
microbes in the periphery, by Trojan Horse mechanisms
(Toborek et al., 2005; Pulzova et al., 2009; Elsheikha and
Khan, 2010; Kaushik et al., 2011). Once inside the CNS,
the neurons, microglia or astrocytes can be infected or
activated by microbial toxins, such as LPS. Innate immu-
nity plays an important role here, as the microbial ligands
are sensed by toll-like receptors (TLRs) leading to signal
transduction via NFkB. The subsequent release of
(inflammatory) mediators by the activated astroglia might
then alter neurological function and affect BBB function
and integrity (Kang and McGavern, 2010).
Remarkably, not all pathogens that cause neurological
disease breach the vascular barrier and enter the CNS. In
low numbers, circulating microbes and their toxins might
not penetrate into the CNS but can still activate the
BBB-EC to release signals that affect the underlying
neuropil (i.e. brain cells) (Fig. 2). Mild stimulation of the
brain endothelium with low amounts of microbes/toxins
increases expression of cell adhesion molecules and
release of cytokines/chemokines, whereas stronger
stimulation also includes opening of the intercellular junc-
tions and increased barrier permeability (Tripathi et al.,
2006; 2009), thus exposing the brain to neurotoxic plasma
substances, including serum albumin that can induce
seizures (Friedman et al., 2009; Hooper et al., 2009) or
antibodies to ion channels, thus affecting neurological
function (Lang et al., 2005). Little is known how brain EC
activation by itself can contribute to neuropathogenesis.
Here, we examine microbial CNS infections in the context
of the BBB as a relay station and modulator of astroglial–
neuronal function.
A fascinating example of peripheral influences that
affect neurological functioning can be appreciated in cere-
bral malaria (CM) where the Plasmodium falciparum para-
How can the blood–brain barrier endothelium influence the brain? 1471
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
site that lives inside an infected RBC (PRBC) ‘remodels’
the PRBC surface with parasite-encoded proteins (Pf-
EMP-1) that then bind to endothelial receptors such as
ICAM-1, resulting in a sequestration of PRBCs in the
vascular lumen. PRBC sequestration is considered
central to CM pathology and concomitant with other brain-
endothelial activating factors likely contributes to the neu-
rological symptoms (Turner et al., 1994; Adams et al.,
2002; van der Heyde et al., 2006; Combes et al., 2010).
Non-immune adults (e.g. travellers) and mostly young
children are especially susceptible to CM, manifested by
impaired consciousness, seizures and coma (Idro et al.,
2005). Unfortunately, even after successful treatment and
recovery, neurological sequelae can remain (Idro et al.,
2010). Understanding CM pathogenesis is therefore
of utmost importance and may clarify the BBB role in
other neurological diseases. In our laboratories we are
currently studying the mechanisms whereby PRBC
induce neurological dysfunction. Using Transwelltm
-based
in vitro human BBB models validated for CM research
(Tripathi et al., 2006; 2007; 2009), we found that PRBC-
mediated activation of the host BBB-EC leads to increase
expression of luminal ICAM-1 and polarized release of
cytokines/chemokine to both luminal (blood-facing) side
and basal (brain-facing) side of the monolayers. A signifi-
cant loss of barrier integrity was only observed upon addi-
tion of high numbers of PRBC. Traditionally cytokines/
chemokines are thought to be released to the luminal side
of the BBB in order to alarm the immune system.
However, circulating cytokines can also shuttle across the
BBB into the CNS (Banks et al., 1995) and both luminal
and basal release of cytokines from BBB-ECs in response
to LPS has also been described (Verma et al., 2006). To
test if the BBB secretions towards the brain side would
affect astrocytes and/or neurons, we collected this con-
ditioned BBB medium from our CM experiments and
added it to astrocyte and neuronal cultures (Fig. 1). Here,
we found a concentration-dependent activation of astro-
cytes (by GFAP immunocytochemistry) and in neurons
disruption of axonal transport and retraction of neuronal
extensions (Stins laboratory, unpubl. obs.). This is in
agreement with axonal damage as found in human
CM autopsies (Medana et al., 2007). These cytokines/
chemokines present in the basal BBB-EC secretions may
play a role in glial cell migration, angiogenesis, tumorigen-
esis, and wound healing, but also modulate astrocyte–
neuronal interactions and thus be involved in neurological
dysfunction.
BBB activation by other pathogens
In advanced stages of Toxoplasma gondii encephalitis
(TE), BALB/c and more sensitive C57BL/6 murine models
show BBB activation and damage as reflected by (i) a
prominent induction of major histocompatibility complex
(MHC) class I and II antigens, (ii) cell adhesion molecules
VCAM-1 and ICAM-1 expression (Deckert-Schluter et al.,
1999; Wang et al., 2007), (iii) a higher ALCAM expression
and (iv) increased BBB permeability (Silva et al., 2010).
T. gondii binds and infects retinal-brain endothelial cells,
bovine umbilical vein endothelial cells (BUVEC) and
human endothelium in vitro, leading to BBB activation. In
retinal EC and BUVEC, T. gondii induces increases in
transcripts for E-, P-selectin, VCAM-1 and ICAM-1 within
1–4 h and inflammatory mediators including GRO1,
MCP-1, Fractalkine, RANTES, IL8, IP10, MCP-1,
GM-CSF, COX2 and iNOS persist at least up to 72 h post
infection (Daubener et al., 2001; Stumbo et al., 2002;
Fig. 1. In vitro set-up to study effects of brain endothelium on underlying brain cells. Shown here is a Transwelltm
tissue culture insert that can
be seeded with brain endothelial cells and inserted in a tissue culture wells, thus creating a two-chamber model where the upper compartment
represents the blood side and the bottom compartment the brain side. Dependent on the experimental set-up astrocytes, pericytes can be
incorporated on the abluminal side on the membrane (in contact with the brain endothelial cells) (B), in the bottom compartment or in a
separate tissue culture well (A). Upon confluence and when sufficient barrier function is obtained, microbes can be added to the upper
compartment. Activation of the BBB endothelium, release of factors to either side and their effects on the underlying brain cells can then be
studied.
1472 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
Smith et al., 2004; Knight et al., 2005; Taubert et al.,
2006a,b). Once T. gondii crosses the BBB, subsequent
microglial infection and astrocyte activation can then have
a negative effect on the BBB leading to activation and loss
of integrity (Silva et al., 2010; Dellacasa-Lindberg et al.,
2011).
African trypanosomes (Trypanosoma brucei spp.)
upregulate brain endothelial expression of ICAM-1,
VCAM-1 and E-selectin and release of IL-6, CXCL8,
CCL2 and TNF-a (Mulenga et al., 2001; Girard et al.,
2005; Grab and Kennedy, 2008). Matrix metalloprotease
MMP2 was increased in CSF (Hainard et al., 2011) and in
vitro studies showed that brain endothelium can poten-
tially contribute to this (D. Grab, unpublished). In vitro and
gene-profiling studies showed that Trypanosome transmi-
gration requires cysteine proteases Gaq-mediated
calcium and protease-activated receptor-2 (PAR-2) sig-
nalling (Nikolskaia et al., 2006; Grab et al., 2009a), initial
events that are precursory to CNS neuroinflammatory
disease (Grab et al., 2009a).
Activation of BBB-EC by Borreliia burgdorferi alone
or in co-infection with Anaplasma phygocytophilum-
infected neutrophils has also been observed. Increased
expression of host cell adhesion molecules, plasmino-
gen activators, plasminogen activator receptors and
increases release of cytokines, chemokines and metal-
loproteases might contribute to neuropathogenesis
(Sellati et al., 1995; Burns et al., 1997; Grab et al., 2005;
2007; 2009b).
Blood–brain barrier breakdown as a loss of junctional
molecules has been shown during advanced neuro-HIV
infection based on biopsy and post-mortem samples and
in vivo disease models (Persidsky et al., 2006; van
Horssen et al., 2007). During initial infection, high viral
titres and secreted viral proteins activate the BBB-EC
resulting in increases in expression of cell adhesion mol-
ecules and release of cytokines and chemokines (Stins
et al., 2001b; 2004; Persidsky et al., 2006; Shiu et al.,
2007; Chaudhuri et al., 2008), which directed towards the
brain side can affect neurological functioning and contrib-
utes to sickness behaviour. The dysregulation of the BBB
during and after neuroinvasion is a critical component of
the neuropathogenic process (Strazza et al., 2011). An
indirect pathogen-induced peripheral stimulus can also
alter neurological function and behaviour. Circulating
LPS, which is present in low concentrations in conditions
including HIV infection, alcohol abuse and some major
depression disorders (Frank et al., 2004; Maes et al.,
2008; Nowroozalizadeh et al., 2010), does not readily
enter the CNS, but accumulates in the BBB-EC, but still
astroglial activation is observed (Singh and Jiang, 2004;
Banks and Robinson, 2009).
Mechanisms for BBB-related neurological
modulation
In neurotropic infections, generally similar host responses
are observed at the BBB, but a role of the BBB in confer-
ring neurological dysfunction is most clearly observed in
CM. However, the mechanisms that are responsible are
unclear, as is their exact function and how they will affect
neuronal function. Reduced release of neuroprotective
Fig. 2. Transmission of astroglial and
neuronal modulating signals from the BBB
endothelium. Neurotropic microbes, like
PRBC, HIV-1 or Trypanosomes, can stimulate
the release of soluble factors (represented by
the arrows), such as astroglial-activating
matrix metalloproteases, cytokines and
chemokines such as IL-1b, IL-6, CCL20,
GRO’s and TNFa from the basal side of brain
endothelial cells. Targets for these mediators
are present in the brain on either astroglial
cells and/or neurons and can result in
modulation of astroglial activation and both
directly and indirectly of neuronal function.
Vessel lumen
Microbial ac!va!on
= Cell adhesion
molecules
= signaling
BBB-
endothelium
Brain parenchyma
endothelium
NO
+ ?
MMP’s
Cytokines
Chemokines
Astrocytes/
microglia
+ -? + -?
Neuron
+ -?
Neurological Modula!on > BeneÞts versus Complica!ons
How can the blood–brain barrier endothelium influence the brain? 1473
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
secretions like brain-derived neurotrophic factor (BDNF)
versus increases in release of nitric oxide (NO), glycine,
cytokines, chemokines and/or MMPs could contribute to
these effects (Fig. 2). Dependent on the individual,
release of BBB-EC-derived cytokines/chemokines could
be very robust in CM (Tripathi et al., 2009; Wassmer et al.,
2011). Inside the CNS, the BBB-derived cytokines/
chemokines may play a role in glial cell migration, angio-
genesis, tumorigenesis and wound healing, but may
also modulate astrocyte–neuronal interactions and thus
be involved in neurological dysfunction. At this moment, it
is unclear as to which particular cytokines/chemokines
are involved, their exact function, how they will affect
neuronal functioning and whether their targets are on
brain cells.
Using in vitro BBB-CM models we observed large
increases in CCL20 transcripts and release (Tripathi
et al., 2009). The non-promiscuous receptor for CCL20,
CCR6 is generally non-detectable in control brains,
but upregulated in brain inflammation, such as in the
murine model for CM that replicates certain features of
human pathology (Hunt et al., 2010), and we see
increased CCR6 immunostaining in cells resembling
neuroglia, possible oligodendrocytes. Oligodendrocytes
are believed to be involved in neuronal signal modula-
tion, and as such, BBB-released CCL20 could affect
neuronal transmission. Astrocytes also secrete CXCL1,
2, 3 and their receptor (CXCR2) is present on neuron
(Dorf et al., 2000). CXCR2 can regulate APMA type
glutamate receptor function by increasing receptor coop-
erativety and postsynaptic transmission amplitude (Lax
et al., 2002). Furthermore, IL8 and CXCL1 can modulate
Purkinje neuron activity (Giovannelli et al., 1998).
CXCL1 increases ERK phosphorylation in cortical
neurons (Xia and Hyman, 2002). Thus, these may be
potential mechanisms whereby release of CXCL1, 2 or 3
from the basolateral side of the BBB, may modulate
astroglial and/or neuronal function. Taken together, alter-
ation of the chemokine/receptor ratio in the brain may be
a mechanism to modulate neuronal function, but when
the BBB is over activated, this may lead to neuronal
dysfunction.
Other factors, such as MMPs or NO basally released
from an activated BBB can modulate not only matrix–
BBB-EC interactions and thus barrier integrity but
also synaptic function and long-term potentiation by
cleavage of NR1 subunit of the NMDA receptor and
ICAM-5 (Szklarczyk et al., 2002; Huang et al., 2008;
Conant et al., 2010; Louboutin et al., 2010). In CM, NO
released from the BBB can have a dual effect; luminal
released NO is quenched by free haemoglobin and
reactive oxygen species and therefore results in a low
peripheral NO bioavailability (Gramaglia, 2006). As
serum arginine levels are coupled to eNOS activity, it is
not clear how this affects NO released to the basal side
of the BBB in CM and other neuromicrobial diseases.
NO has various effects on neuronal function, including
anterograde signalling (Fernandez-Alvarez et al., 2011),
cognitive learning (Paul and Ekambaram, 2011), depres-
sion behaviour (Workman et al., 2011), glutamatergic
transmission (Sardo et al., 2011) and altering of sensi-
tivity of dopamine release to presynaptic activation
(Hartung et al., 2011). In astrocytes, NO alters connex-
ins leading to reduction of inter-astrocytic gap-junction
communications (Ball et al., 2011) which could also
affect astrocyte–BBB-EC or astrocyte–neurons com-
munications. Reactive nitrogen species can trigger lipid
peroxidation chain reactions throughout the cerebral
vasculature, and brain parenchyma and thus contribut-
ing neurocognitive sequelae as seen in sepsis survivors
(Berg et al., 2011).
Conclusions
It makes sense that, in the context of the NVU principle,
an alteration in the ‘resting’ status of the BBB-EC by
neurotropic microbes can lead to modulation of
astroglial/neuronal function. Thus, an activated BBB
could function as a communication-relay station between
peripheral signals to the brain and contribute to symp-
toms ranging from mild symptoms such as malaise to
serious neurological dysfunction such as coma in CM.
Brain endothelial-derived modulating factors that play a
role in NVU function include the and increased release
of local and brain-directed cytokines, chemokines, NO
and MMPs versus a decreased release of neurotrophic
and protective factors. It is not clear at what point the
altered release of these factors would tip the balance
from playing a beneficial role in astroneuronal homeo-
stasis and repair to a detrimental one leading to neuro-
logical dysfunction (Fig. 3). Therefore, it is important to
understand BBB-EC factors that are involved in mediat-
ing and regulating the responses to microbial exposure
and ‘decide’ at which point the balance tips from a ben-
eficial neuroprotective response to a detrimental reac-
tion. BBB responses, like in CM or stroke, can be very
local and affecting only small brain areas. Therefore,
changes in these factors can be missed in systemic
measurements of blood or CSF samples. In vitro
systems can aid to identify these factors that subse-
quently can be validated in vivo. Many questions still
remain and further clarification of interactions between
the BBB-EC and underlying CNS components are
needed as this may not only lead to novel therapeutic
targets for treating neurological dysfunction in a wide
variety of disorders but could also be beneficial in the
repair of astroneuronal function in conditions such as
stroke.
1474 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
References
Abbott, N.J., Ronnback, L., and Hansson, E. (2006)
Astrocyte-endothelial interactions at the blood–brain
barrier. Nat Rev Neurosci 7: 41–53.
Adams, S., Brown, H., and Turner, G. (2002) Breaking down
the blood–brain barrier: signaling a path to cerebral
malaria? Trends Parasitol 18: 360–366.
Ball, K.K., Harik, L., Gandhi, G.K., Cruz, N.F., and Dienel,
G.A. (2011) Reduced gap junctional communication among
astrocytes in experimental diabetes: contributions of
altered connexin protein levels and oxidative-nitrosative
modifications. J Neurosci Res doi: 10.1002/jnr.22663
Banks, W.A., and Robinson, S.M. (2009) Minimal penetration
of lipopolysaccharide across the murine blood–brain
barrier. Brain Behav Immun 24: 102–109.
Banks, W.A., Kastin, A.J., and Broadwell, R.D. (1995)
Passage of cytokines across the blood–brain barrier. Neu-
roimmunomodulation 2: 241–248.
Barth, M.C., Ahluwalia, N., Anderson, T.J., Hardy, G.J.,
Sinha, S., Alvarez-Cardona, J.A., et al. (2009) Kynurenic
acid triggers firm arrest of leukocytes to vascular endothe-
lium under flow conditions. J Biol Chem 284: 19189–
19195.
Berg, R.M., Moller, K., and Bailey, D.M. (2011) Neuro-
oxidative-nitrosative stress in sepsis. J Cereb Blood Flow
Metab 31: 1532–1544.
Bonkowski, D., Katyshev, V., Balabanov, R.D., Borisov, A.,
and Dore-Duffy, P. (2011) The CNS microvascular pericyte:
pericyte-astrocyte crosstalk in the regulation of tissue sur-
vival. Fluids Barriers CNS 8: 8.
Burns, M.J., Sellati, T.J., Teng, E.I., and Furie, M.B. (1997)
Production of interleukin-8 (IL-8) by cultured endothelial
cells in response to Borrelia burgdorferi occurs indepen-
dently of secreted [corrected] IL-1 and tumor necrosis
factor alpha and is required for subsequent transendothe-
lial migration of neutrophils. Infect Immun 65: 1217–1222.
Chaudhuri, A., Duan, F., Morsey, B., Persidsky, Y., and
Kanmogne, G.D. (2008) HIV-1 activates proinflammatory
and interferon-inducible genes in human brain microvascu-
lar endothelial cells: putative mechanisms of blood–brain
barrier dysfunction. J Cereb Blood Flow Metab 28: 697–
711.
Chintawar, S., Cayrol, R., Antel, J., Pandolfo, M., and Prat, A.
(2009) Blood–brain barrier promotes differentiation of
human fetal neural precursor cells. Stem Cells 27: 838–846.
Chung, Y.C., Ko, H.W., Bok, E., Park, E.S., Huh, S.H., Nam,
J.H., et al. (2010) The role of neuroinflammation on the
pathogenesis of Parkinson’s disease. BMB Rep 43: 225–
232.
Combes, V., El-Assaad, F., Faille, D., Jambou, R., Hunt, N.H.,
and Grau, G.E. (2010) Microvesiculation and cell interac-
tions at the brain–endothelial interface in cerebral malaria
pathogenesis. Prog Neurobiol 91: 140–151.
Conant, K., Wang, Y., Szklarczyk, A., Dudak, A., Mattson,
M.P., and Lim, S.T. (2010) Matrix metalloproteinase-
dependent shedding of intercellular adhesion molecule-5
occurs with long-term potentiation. Neuroscience 166:
508–521.
Cucullo, L., Hossain, M., Puvenna, V., Marchi, N., and
Janigro, D. (2011a) The role of shear stress in Blood–Brain
Barrier endothelial physiology. BMC Neurosci 12: 40.
Cucullo, L., Marchi, N., Hossain, M., and Janigro, D. (2011b)
A dynamic in vitro BBB model for the study of immune cell
trafficking into the central nervous system. J Cereb Blood
Flow Metab 31: 767–777.
Cunningham, C., Campion, S., Lunnon, K., Murray, C.L.,
Woods, J.F., Deacon, R.M., et al. (2009) Systemic inflam-
mation induces acute behavioral and cognitive changes
and accelerates neurodegenerative disease. Biol Psychia-
try 65: 304–312.
Daubener, W., Spors, B., Hucke, C., Adam, R., Stins, M.,
Kim, K.S., et al. (2001) Restriction of Toxoplasma gondii
growth in human brain microvascular endothelial cells by
activation of indoleamine 2,3-dioxygenase. Infect Immun
69: 6527–6531.
Deckert-Schluter, M., Bluethmann, H., Kaefer, N., Rang, A.,
and Schluter, D. (1999) Interferon-gamma receptor-
mediated but not tumor necrosis factor receptor type 1- or
type 2-mediated signaling is crucial for the activation of
Fig. 3. The balance of factors derived from the activated BBB
endothelium could determine a neuroprotective versus detrimental
response. Under homeostatic conditions, the BBB endothelium
releases substances such as cytokines, chemokines, NO and
MMPs, neurotrophic and neuroprotective factors towards the brain
side. Upon activation, the BBB endothelium releases factors in
order to ‘alert’ brain cells and protect the neurological function.
However, with strong activation the BBB may ‘overreact’ and these
otherwise positive factors may turn into detrimental substances. A
decrease in neuroprotective and neurotrophic secretions may occur
concomitantly. It is not yet clear which BBB-EC factors govern the
release of these substances and at what point the balance tips
from being protective of the brain’s homeostasis to being negative
and affecting astroneuronal function.
How can the blood–brain barrier endothelium influence the brain? 1475
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
cerebral blood vessel endothelial cells and microglia in
murine Toxoplasma encephalitis. Am J Pathol 154: 1549–
1561.
Dellacasa-Lindberg, I., Fuks, J.M., Arrighi, R.B., Lambert, H.,
Wallin, R.P., Chambers, B.J., et al. (2011) Migratory acti-
vation of primary cortical microglia upon infection with
Toxoplasma gondii. Infect Immun 79: 3046–3052.
Dorf, M.E., Berman, M.A., Tanabe, S., Heesen, M., and Luo,
Y. (2000) Astrocytes express functional chemokine recep-
tors. J Neuroimmunol 111: 109–121.
Elsheikha, H.M., and Khan, N.A. (2010) Protozoa traversal of
the blood–brain barrier to invade the central nervous
system. FEMS Microbiol Rev 34: 532–553.
Eugenin, E.A., Osiecki, K., Lopez, L., Goldstein, H.,
Calderon, T.M., and Berman, J.W. (2006) CCL2/monocyte
chemoattractant protein-1 mediates enhanced transmigra-
tion of human immunodeficiency virus (HIV)-infected
leukocytes across the blood–brain barrier: a potential
mechanism of HIV-CNS invasion and NeuroAIDS.
J Neurosci 26: 1098–1106.
Fernandez-Alvarez, A., Gomez-Sena, L., Fabbiani, M.G.,
Budelli, R., and Abudara, V. (2011) Endogenous pre-
synaptic nitric oxide supports an anterograde signaling
in the central nervous system. J Neurochem 118: 546–
557.
Frank, J., Witte, K., Schrodl, W., and Schutt, C. (2004)
Chronic alcoholism causes deleterious conditioning of
innate immunity. Alcohol Alcohol 39: 386–392.
Friedman, A., Kaufer, D., and Heinemann, U. (2009) Blood–
brain barrier breakdown-inducing astrocytic transforma-
tion: novel targets for the prevention of epilepsy. Epilepsy
Res 85: 142–149.
Giovannelli, A., Limatola, C., Ragozzino, D., Mileo, A.M.,
Ruggieri, A., Ciotti, M.T., et al. (1998) CXC chemokines
interleukin-8 (IL-8) and growth-related gene product alpha
(GROalpha) modulate Purkinje neuron activity in mouse
cerebellum. J Neuroimmunol 92: 122–132.
Girard, M., Giraud, S., Courtioux, B., Jauberteau-Marchan,
M.O., and Bouteille, B. (2005) Endothelial cell activation in
the presence of African trypanosomes. Mol Biochem Para-
sitol 139: 41–49.
Grab, D.J., and Kennedy, P.G. (2008) Traversal of human
and animal trypanosomes across the blood–brain barrier.
J Neurovirol 14: 344–351.
Grab, D.J., Perides, G., Dumler, J.S., Kim, K.J., Park, J., Kim,
Y.V., et al. (2005) Borrelia burgdorferi, host-derived pro-
teases, and the blood–brain barrier. Infect Immun 73:
1014–1022.
Grab, D.J., Nyarko, E., Barat, N.C., Nikolskaia, O.V., and
Dumler, J.S. (2007) Anaplasma phagocytophilum–Borrelia
burgdorferi coinfection enhances chemokine, cytokine, and
matrix metalloprotease expression by human brain
microvascular endothelial cells. Clin Vaccine Immunol 14:
1420–1424.
Grab, D.J., Garcia-Garcia, J.C., Nikolskaia, O.V., Kim, Y.V.,
Brown, A., Pardo, C.A., et al. (2009a) Protease activated
receptor signaling is required for African trypanosome tra-
versal of human brain microvascular endothelial cells.
PLoS Negl Trop Dis 3: e479.
Grab, D.J., Nyarko, E., Nikolskaia, O.V., Kim, Y.V., and
Dumler, J.S. (2009b) Human brain microvascular endo-
thelial cell traversal by Borrelia burgdorferi requires
calcium signaling. Clin Microbiol Infect 15: 422–426.
Gramaglia, I. Low nitric oxide bioavailability contributes to the
genesis of experimental cerebral malaria. Sobolewski, P.,
Meays, D., Contreras, R., Nolan, J.P., Frangos, J.A., Inta-
glietta, M., and van der Heyde, H.C. Nat Med 12 [12],
1417–1422. 2006. Ref Type: Generic.
Guo, S., Kim, W.J., Lok, J., Lee, S.R., Besancon, E., Luo,
B.H., et al. (2008) Neuroprotection via matrix-trophic cou-
pling between cerebral endothelial cells and neurons. Proc
Natl Acad Sci USA 105: 7582–7587.
Hainard, A., Tiberti, N., Robin, X., Ngoyi, D.M., Matovu, E.,
Enyaru, J.C., et al. (2011) Matrix metalloproteinase-9 and
intercellular adhesion molecule 1 are powerful staging
markers for human African trypanosomiasis. Trop Med Int
Health 16: 119–126.
Hartung, H., Threlfell, S., and Cragg, S.J. (2011) Nitric oxide
donors enhance the frequency dependence of dopamine
release in nucleus accumbens. Neuropsychopharmacol-
ogy 36: 1811–1822.
Hatherell, K., Couraud, P.O., Romero, I.A., Weksler, B., and
Pilkington, G.J. (2011) Development of a three-
dimensional, all-human in vitro model of the blood–brain
barrier using mono-, co-, and tri-cultivation Transwell
models. J Neurosci Methods 199: 223–229.
van der Heyde, H.C., Nolan, J., Combes, V., Gramaglia, I.,
and Grau, G.E. (2006) A unified hypothesis for the genesis
of cerebral malaria: sequestration, inflammation and hemo-
stasis leading to microcirculatory dysfunction. Trends Para-
sitol 22: 503–508.
Holman, D.W., Klein, R.S., and Ransohoff, R.M. (2011) The
blood–brain barrier, chemokines and multiple sclerosis.
Biochim Biophys Acta 1812: 220–230.
Hooper, C., Pinteaux-Jones, F., Fry, V.A., Sevastou, I.G.,
Baker, D., Heales, S.J., et al. (2009) Differential effects of
albumin on microglia and macrophages; implications for
neurodegeneration following blood–brain barrier damage.
J Neurochem 109: 694–705.
van Horssen, J., Brink, B.P., de Vries, H.E., van der Valk, P.,
and Bo, L. (2007) The blood–brain barrier in cortical mul-
tiple sclerosis lesions. J Neuropathol Exp Neurol 66: 321–
328.
Huang, W., Rha, G.B., Han, M.J., Eum, S.Y., Andras, I.E.,
Zhong, Y., et al. (2008) PPARalpha and PPARgamma
effectively protect against HIV-induced inflammatory
responses in brain endothelial cells. J Neurochem 107:
497–509.
Humpel, C. (2011) Chronic mild cerebrovascular dysfunction
as a cause for Alzheimer’s disease? Exp Gerontol 46:
225–232.
Hunt, N.H., Grau, G.E., Engwerda, C., Barnum, S.R., van der
Heyde, H., Hansen, D.S., et al. (2010) Murine cerebral
malaria: the whole story. Trends Parasitol 26: 272–
274.
Idro, R., Jenkins, N.E., and Newton, C.R. (2005) Pathogen-
esis, clinical features, and neurological outcome of cere-
bral malaria. Lancet Neurol 4: 827–840.
Idro, R., Kakooza-Mwesige, A., Balyejjussa, S., Mirembe, G.,
Mugasha, C., Tugumisirize, J., et al. (2010) Severe neuro-
logical sequelae and behaviour problems after cerebral
malaria in Ugandan children. BMC Res Notes 3: 104.
1476 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
Kamouchi, M., Ago, T., and Kitazono, T. (2011) Brain peri-
cytes: emerging concepts and functional roles in brain
homeostasis. Cell Mol Neurobiol 31: 175–193.
Kang, S.S., and McGavern, D.B. (2010) Microbial induction of
vascular pathology in the CNS. J Neuroimmune Pharmacol
5: 370–386.
Kaushik, D.K., Gupta, M., and Basu, A. (2011) Microglial
response to viral challenges: every silver lining comes with
a cloud. Front Biosci 17: 2187–2205.
Knight, B.C., Brunton, C.L., Modi, N.C., Wallace, G.R., and
Stanford, M.R. (2005) The effect of Toxoplasma gondii
infection on expression of chemokines by rat retinal vas-
cular endothelial cells. J Neuroimmunol 160: 41–47.
Kroll, S., El-Gindi, J., Thanabalasundaram, G., Panpumthong,
P., Schrot, S., Hartmann, C., et al. (2009) Control
of the blood–brain barrier by glucocorticoids and the cells of
the neurovascular unit. Ann N Y Acad Sci 1165: 228–
239.
Lang, B., Newbold, C.I., Williams, G., Peshu, N., Marsh, K.,
and Newton, C.R. (2005) Antibodies to voltage-gated
calcium channels in children with falciparum malaria.
J Infect Dis 191: 117–121.
Langenkamp, E., and Molema, G. (2009) Microvascular
endothelial cell heterogeneity: general concepts and phar-
macological consequences for anti-angiogenic therapy of
cancer. Cell Tissue Res 335: 205–222.
Lax, P., Limatola, C., Fucile, S., Trettel, F., Di Bartolomeo, S.,
Renzi, M., et al. (2002) Chemokine receptor CXCR2 regu-
lates the functional properties of AMPA-type glutamate
receptor GluR1 in HEK cells. J Neuroimmunol 129: 66–73.
Lok, J., Gupta, P., Guo, S., Kim, W.J., Whalen, M.J., van
Leyen, K., et al. (2007) Cell–cell signaling in the neurovas-
cular unit. Neurochem Res 32: 2032–2045.
Louboutin, J.P., Agrawal, L., Reyes, B.A., Van Bockstaele,
E.J., and Strayer, D.S. (2010) HIV-1 gp120-induced injury
to the blood–brain barrier: role of metalloproteinases 2 and
9 and relationship to oxidative stress. J Neuropathol Exp
Neurol 69: 801–816.
Maes, M., Kubera, M., and Leunis, J.C. (2008) The gut-brain
barrier in major depression: intestinal mucosal dysfunction
with an increased translocation of LPS from gram negative
enterobacteria (leaky gut) plays a role in the inflammatory
pathophysiology of depression. Neuro Endocrinol Lett 29:
117–124.
Medana, I.M., Day, N.P., Hien, T.T., Mai, N.T., Bethell, D.,
Phu, N.H., et al. (2007) Cerebral calpain in fatal falciparum
malaria. Neuropathol Appl Neurobiol 33: 179–192.
Miklossy, J. (2008) Chronic inflammation and amyloidogen-
esis in Alzheimer’s disease – role of Spirochetes.
J Alzheimers Dis 13: 381–391.
Mulenga, C., Mhlanga, J.D., Kristensson, K., and Robertson,
B. (2001) Trypanosoma brucei brucei crosses the blood–
brain barrier while tight junction proteins are preserved in a
rat chronic disease model. Neuropathol Appl Neurobiol 27:
77–85.
Nakagawa, S., Deli, M.A., Kawaguchi, H., Shimizudani, T.,
Shimono, T., Kittel, A., et al. (2009) A new blood–brain
barrier model using primary rat brain endothelial cells, peri-
cytes and astrocytes. Neurochem Int 54: 253–263.
Neuhaus, W., Freidl, M., Szkokan, P., Berger, M., Wirth, M.,
Winkler, J., et al. (2011) Effects of NMDA receptor modu-
lators on a blood–brain barrier in vitro model. Brain Res
1394: 49–61.
Nikolskaia, O.V., de A Lima, A.P., Kim, Y.V., Lonsdale-Eccles,
J.D., Fukuma, T., Scharfstein, J., et al. (2006) Blood–brain
barrier traversal by African trypanosomes requires calcium
signaling induced by parasite cysteine protease. J Clin
Invest 116: 2739–2747.
Nowroozalizadeh, S., Mansson, F., da, S.Z., Repits, J., Dabo,
B., Pereira, C., et al. (2010) Microbial translocation corre-
lates with the severity of both HIV-1 and HIV-2 infections.
J Infect Dis 201: 1150–1154.
Ochoa-Reparaz, J., Mielcarz, D.W., Begum-Haque, S., and
Kasper, L.H. (2011) Gut, bugs, and brain: role of commen-
sal bacteria in the control of central nervous system
disease. Ann Neurol 69: 240–247.
Paul, V., and Ekambaram, P. (2011) Involvement of nitric
oxide in learning & memory processes. Indian J Med Res
133: 471–478.
Persidsky, Y., Ramirez, S.H., Haorah, J., and Kanmogne,
G.D. (2006) Blood–brain barrier: structural components
and function under physiologic and pathologic conditions.
J Neuroimmune Pharmacol 1: 223–236.
Phiri, H., Montgomery, J., Molyneux, M., and Craig, A. (2009)
Competitive endothelial adhesion between Plasmodium
falciparum isolates under physiological flow conditions.
Malar J 8: 214.
Pulzova, L., Bhide, M.R., and Andrej, K. (2009) Pathogen
translocation across the blood–brain barrier. FEMS
Immunol Med Microbiol 57: 203–213.
Reale, M., Iarlori, C., Thomas, A., Gambi, D., Perfetti, B., Di
Nicola, M., et al. (2009) Peripheral cytokines profile in Par-
kinson’s disease. Brain Behav Immun 23: 55–63.
Roux, F., and Couraud, P.O. (2005) Rat brain endothelial cell
lines for the study of blood–brain barrier permeability and
transport functions. Cell Mol Neurobiol 25: 41–58.
Sardo, P., Carletti, F., Rizzo, V., Lonobile, G., Friscia, S., and
Ferraro, G. (2011) Nitric oxide-active compounds modulate
the intensity of glutamate-evoked responses in the globus
pallidus of the rat. Life Sci 88: 1113–1120.
Schafer, C., Parlesak, A., Schutt, C., Bode, J.C., and Bode,
C. (2002) Concentrations of lipopolysaccharide-binding
protein, bactericidal/permeability-increasing protein,
soluble CD14 and plasma lipids in relation to endotox-
aemia in patients with alcoholic liver disease. Alcohol
Alcohol 37: 81–86.
Sellati, T.J., Burns, M.J., Ficazzola, M.A., and Furie, M.B.
(1995) Borrelia burgdorferi upregulates expression of
adhesion molecules on endothelial cells and promotes
transendothelial migration of neutrophils in vitro. Infect
Immun 63: 4439–4447.
Shiu, C., Barbier, E., Di, C.F., Choi, H.J., and Stins, M. (2007)
HIV-1 gp120 as well as alcohol affect blood–brain barrier
permeability and stress fiber formation: involvement of
reactive oxygen species. Alcohol Clin Exp Res 31: 130–
137.
Silva, N.M., Manzan, R.M., Carneiro, W.P., Milanezi, C.M.,
Silva, J.S., Ferro, E.A., et al. (2010) Toxoplasma gondii: the
severity of toxoplasmic encephalitis in C57BL/6 mice is
associated with increased ALCAM and VCAM-1 expres-
sion in the central nervous system and higher blood–brain
barrier permeability. Exp Parasitol 126: 167–177.
How can the blood–brain barrier endothelium influence the brain? 1477
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
Singh, A.K., and Jiang, Y. (2004) How does peripheral
lipopolysaccharide induce gene expression in the brain of
rats? Toxicology 201: 197–207.
Smith, J.R., Franc, D.T., Carter, N.S., Zamora, D., Planck,
S.R., and Rosenbaum, J.T. (2004) Susceptibility of retinal
vascular endothelium to infection with Toxoplasma gondii
tachyzoites. Invest Ophthalmol Vis Sci 45: 1157–1161.
Stins, M.F., Prasadarao, N.V., Zhou, J., Arditi, M., and Kim,
K.S. (1997) Bovine brain microvascular endothelial cells
transfected with SV40-large T antigen: development of an
immortalized cell line to study pathophysiology of CNS
disease. In Vitro Cell Dev Biol Anim 33: 243–247.
Stins, M.F., Badger, J., and Sik Kim, K. (2001a) Bacterial
invasion and transcytosis in transfected human brain
microvascular endothelial cells. Microb Pathog 30: 19–28.
Stins, M.F., Shen, Y., Huang, S.H., Gilles, F., Kalra, V.K., and
Kim, K.S. (2001b) Gp120 activates children’s brain endot-
helial cells via CD4. J Neurovirol 7: 125–134.
Stins, M.F., Pearce, D., Choi, H., Di Cello, F., Pardo, C.A.,
and Kim, K.S. (2004) CD4 and chemokine receptors on
human brain microvascular endothelial cells, implications
for human immunodeficiency virus type 1 pathogenesis.
Endothelium 11: 275–284.
Strazza, M., Pirrone, V., Wigdahl, B., and Nonnemacher,
M.R. (2011) Breaking down the barrier: the effects of HIV-1
on the blood–brain barrier. Brain Res 5: 1396–1399.
Stumbo, A.C., Barbosa, H.S., Carvalho, T.M., Porto, L.C.,
and Carvalho, L. (2002) Anionic sites, fucose residues and
class I human leukocyte antigen fate during interaction of
Toxoplasma gondii with endothelial cells. Mem Inst
Oswaldo Cruz 97: 517–522.
Szklarczyk, A., Lapinska, J., Rylski, M., McKay, R.D., and
Kaczmarek, L. (2002) Matrix metalloproteinase-9 under-
goes expression and activation during dendritic remodeling
in adult hippocampus. J Neurosci 22: 920–930.
Tan, K.H., Dobbie, M.S., Felix, R.A., Barrand, M.A., and
Hurst, R.D. (2001) A comparison of the induction of immor-
talized endothelial cell impermeability by astrocytes. Neu-
roreport 12: 1329–1334.
Taubert, A., Krull, M., Zahner, H., and Hermosilla, C. (2006a)
Toxoplasma gondii and Neospora caninum infections of
bovine endothelial cells induce endothelial adhesion mol-
ecule gene transcription and subsequent PMN adhesion.
Vet Immunol Immunopathol 112: 272–283.
Taubert, A., Zahner, H., and Hermosilla, C. (2006b) Dynam-
ics of transcription of immunomodulatory genes in endot-
helial cells infected with different coccidian parasites. Vet
Parasitol 142: 214–222.
Toborek, M., Lee, Y.W., Flora, G., Pu, H., Andras, I.E., Wyle-
gala, E., et al. (2005) Mechanisms of the blood–brain
barrier disruption in HIV-1 infection. Cell Mol Neurobiol 25:
181–199.
Tripathi, A.K., Sullivan, D.J., and Stins, M.F. (2006) Plasmo-
dium falciparum-infected erythrocytes increase intercellu-
lar adhesion molecule 1 expression on brain endothelium
through NF-kappaB. Infect Immun 74: 3262–3270.
Tripathi, A.K., Sullivan, D.J., and Stins, M.F. (2007) Plasmo-
dium falciparum-infected erythrocytes decrease the integ-
rity of human blood–brain barrier endothelial cell
monolayers. J Infect Dis 195: 942–950.
Tripathi, A.K., Sha, W., Shulaev, V., Stins, M.F., and Sullivan,
D.J., Jr (2009) Plasmodium falciparum-infected erythro-
cytes induce NF-kappaB regulated inflammatory pathways
in human cerebral endothelium. Blood 114: 4243–4252.
Turner, G.D., Morrison, H., Jones, M., Davis, T.M., Looaree-
suwan, S., Buley, I.D., et al. (1994) An immunohistochemi-
cal study of the pathology of fatal malaria. Evidence for
widespread endothelial activation and a potential role for
intercellular adhesion molecule-1 in cerebral sequestra-
tion. Am J Pathol 145: 1057–1069.
Verma, S., Nakaoke, R., Dohgu, S., and Banks, W.A. (2006)
Release of cytokines by brain endothelial cells: a polarized
response to lipopolysaccharide. Brain Behav Immun 20:
449–455.
Wang, X., Michie, S.A., Xu, B., and Suzuki, Y. (2007) Impor-
tance of IFN-gamma-mediated expression of endothelial
VCAM-1 on recruitment of CD8+ T cells into the brain
during chronic infection with Toxoplasma gondii.
J Interferon Cytokine Res 27: 329–338.
Wassmer, S.C., Moxon, C.A., Taylor, T., Grau, G.E.,
Molyneux, M.E., and Craig, A.G. (2011) Vascular endothe-
lial cells cultured from patients with cerebral or uncompli-
cated malaria exhibit differential reactivity to TNF. Cell
Microbiol 13: 198–209.
Weiss, J.M., Nath, A., Major, E.O., and Berman, J.W. (1999)
HIV-1 Tat induces monocyte chemoattractant protein-1-
mediated monocyte transmigration across a model of the
human blood–brain barrier and up-regulates CCR5 expres-
sion on human monocytes. J Immunol 163: 2953–2959.
Weksler, B.B., Subileau, E.A., Perriere, N., Charneau, P.,
Holloway, K., Leveque, M., et al. (2005) Blood–brain
barrier-specific properties of a human adult brain endothe-
lial cell line. FASEB J 19: 1872–1874.
Workman, J.L., Weber, M.D., and Nelson, R.J. (2011) Dietary
arginine depletion reduces depressive-like responses in
male, but not female, mice. Behav Brain Res 223: 81–
87.
Xia, M., and Hyman, B.T. (2002) GROalpha/KC, a chemokine
receptor CXCR2 ligand, can be a potent trigger for neu-
ronal ERK1/2 and PI-3 kinase pathways and for tau
hyperphosphorylation-a role in Alzheimer’s disease?
J Neuroimmunol 122: 55–64.
Zhang, Y., and Barres, B.A. (2010) Astrocyte heterogeneity:
an underappreciated topic in neurobiology. Curr Opin Neu-
robiol 20: 588–594.
Zhang, R., Miller, R.G., Gascon, R., Champion, S., Katz, J.,
Lancero, M., et al. (2009) Circulating endotoxin and sys-
temic immune activation in sporadic amyotrophic lateral
sclerosis (sALS). J Neuroimmunol 206: 121–124.
Zlokovic, B.V. (2008) The blood–brain barrier in health and
chronic neurodegenerative disorders. Neuron 57: 178–
201.
1478 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins
© 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478

More Related Content

What's hot

Modulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticModulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticYildiz Kelahmetoglu
 
Proteínas en Medicina
Proteínas en MedicinaProteínas en Medicina
Proteínas en MedicinaCamila Ospina
 
Brain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainBrain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainRalph Sherman
 
Clinical Neurology and Neurosurgery
Clinical Neurology and NeurosurgeryClinical Neurology and Neurosurgery
Clinical Neurology and NeurosurgeryKaren Reyes
 
PPT Differentiation_of_neural_cells_in_human_embryonic_stem
PPT Differentiation_of_neural_cells_in_human_embryonic_stemPPT Differentiation_of_neural_cells_in_human_embryonic_stem
PPT Differentiation_of_neural_cells_in_human_embryonic_stemCheng Han-Ni
 
Seminario Biologia Molecular
Seminario Biologia Molecular Seminario Biologia Molecular
Seminario Biologia Molecular katerinaruiz
 
Christian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationChristian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationbestwebsite2008
 
Stem Cell Therapy of Spinal Cord Injury
Stem Cell Therapy of Spinal Cord InjuryStem Cell Therapy of Spinal Cord Injury
Stem Cell Therapy of Spinal Cord Injurywiseyou
 
Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Ewelina Maliszewska-Cyna, PhD
 
Stem cell research in Parkinson's Disease
Stem cell research  in Parkinson's DiseaseStem cell research  in Parkinson's Disease
Stem cell research in Parkinson's DiseaseVeena Shriram
 
Neuronal and glial differentiation of human pluripotent stem cells
Neuronal and glial differentiation of human pluripotent stem cellsNeuronal and glial differentiation of human pluripotent stem cells
Neuronal and glial differentiation of human pluripotent stem cellsDiana Santos
 
Stem Cell Treatment At Tiantan Puhua Hospital 4
Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4
Stem Cell Treatment At Tiantan Puhua Hospital 4randyrobinsonpuhua
 
CiteXplore - a more sophisticated search in the life science literature
CiteXplore - a more sophisticated search in the life science literatureCiteXplore - a more sophisticated search in the life science literature
CiteXplore - a more sophisticated search in the life science literatureNikolay12
 
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...johnohab
 

What's hot (19)

Stem cells & Neurodegenerative diseases And Some clinical CNS
Stem cells & Neurodegenerative diseases And Some clinical CNSStem cells & Neurodegenerative diseases And Some clinical CNS
Stem cells & Neurodegenerative diseases And Some clinical CNS
 
The brain and energy
The brain and energyThe brain and energy
The brain and energy
 
Modulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeuticModulation_of_autophagy_as_a_therapeutic
Modulation_of_autophagy_as_a_therapeutic
 
Stem cell Biobridge
Stem cell BiobridgeStem cell Biobridge
Stem cell Biobridge
 
Proteínas en Medicina
Proteínas en MedicinaProteínas en Medicina
Proteínas en Medicina
 
Brain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spainBrain metabolism, eeg, 2012 case study from madrid spain
Brain metabolism, eeg, 2012 case study from madrid spain
 
Clinical Neurology and Neurosurgery
Clinical Neurology and NeurosurgeryClinical Neurology and Neurosurgery
Clinical Neurology and Neurosurgery
 
PPT Differentiation_of_neural_cells_in_human_embryonic_stem
PPT Differentiation_of_neural_cells_in_human_embryonic_stemPPT Differentiation_of_neural_cells_in_human_embryonic_stem
PPT Differentiation_of_neural_cells_in_human_embryonic_stem
 
Seminario Biologia Molecular
Seminario Biologia Molecular Seminario Biologia Molecular
Seminario Biologia Molecular
 
Christian drapeau theory_of_regeneration
Christian drapeau theory_of_regenerationChristian drapeau theory_of_regeneration
Christian drapeau theory_of_regeneration
 
Stem Cell Therapy of Spinal Cord Injury
Stem Cell Therapy of Spinal Cord InjuryStem Cell Therapy of Spinal Cord Injury
Stem Cell Therapy of Spinal Cord Injury
 
Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...Development and implementation of a novel interactome platform in studies on ...
Development and implementation of a novel interactome platform in studies on ...
 
Stem cell research in Parkinson's Disease
Stem cell research  in Parkinson's DiseaseStem cell research  in Parkinson's Disease
Stem cell research in Parkinson's Disease
 
Neuronal and glial differentiation of human pluripotent stem cells
Neuronal and glial differentiation of human pluripotent stem cellsNeuronal and glial differentiation of human pluripotent stem cells
Neuronal and glial differentiation of human pluripotent stem cells
 
Stem Cell Treatment At Tiantan Puhua Hospital 4
Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4Stem  Cell  Treatment At  Tiantan  Puhua  Hospital  4
Stem Cell Treatment At Tiantan Puhua Hospital 4
 
CiteXplore - a more sophisticated search in the life science literature
CiteXplore - a more sophisticated search in the life science literatureCiteXplore - a more sophisticated search in the life science literature
CiteXplore - a more sophisticated search in the life science literature
 
Diapositivas seminario biomol
Diapositivas seminario biomolDiapositivas seminario biomol
Diapositivas seminario biomol
 
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...
Pten Deletion in Adult Neural Stem/Progenitor Cells Enhances Constitutive Neu...
 
Edgardo Arroyo CV
Edgardo Arroyo CVEdgardo Arroyo CV
Edgardo Arroyo CV
 

Similar to Grab Chakravorty Heyde and STINS microbial interactions neuronal function 2011

blood - brain barrier
blood - brain barrierblood - brain barrier
blood - brain barrieralaaaltaee3
 
ChowetalTINSreview.pdf
ChowetalTINSreview.pdfChowetalTINSreview.pdf
ChowetalTINSreview.pdfalaaaltaee3
 
Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Unalissa Freso
 
Drug delivery across BBB
Drug delivery across BBBDrug delivery across BBB
Drug delivery across BBBAman Kumar Naik
 
ijms-23-15271.pdf
ijms-23-15271.pdfijms-23-15271.pdf
ijms-23-15271.pdfalaaaltaee3
 
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdfalaaaltaee3
 
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...Aslam Akhtar, MS
 
PIIS0166223620302861.pdf
PIIS0166223620302861.pdfPIIS0166223620302861.pdf
PIIS0166223620302861.pdfalaaaltaee3
 
Neuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into MemoriesNeuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into Memoriesmustafa sarac
 
Muthusamy_et_al-2015-Aging_Cell
Muthusamy_et_al-2015-Aging_CellMuthusamy_et_al-2015-Aging_Cell
Muthusamy_et_al-2015-Aging_CellLaura Sommerville
 
C-CEBH__ Current Trainees_2015
C-CEBH__ Current Trainees_2015C-CEBH__ Current Trainees_2015
C-CEBH__ Current Trainees_2015Aminah Sheikh
 
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers Cns
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers CnsPharmacopsychiatry Bmsystems Max Planck Publication Biomarkers Cns
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers CnsManuel GEA - Bio-Modeling Systems
 
Cell-Replacement Therapy with Stem Cells in Neurodegenerative Diseases
Cell-Replacement Therapy with Stem Cells in Neurodegenerative DiseasesCell-Replacement Therapy with Stem Cells in Neurodegenerative Diseases
Cell-Replacement Therapy with Stem Cells in Neurodegenerative DiseasesSararajputsa
 
Research article the turing machine theory for some spinal cord and brain con...
Research article the turing machine theory for some spinal cord and brain con...Research article the turing machine theory for some spinal cord and brain con...
Research article the turing machine theory for some spinal cord and brain con...M. Luisetto Pharm.D.Spec. Pharmacology
 
B978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfB978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfsuresh163251
 

Similar to Grab Chakravorty Heyde and STINS microbial interactions neuronal function 2011 (20)

blood - brain barrier
blood - brain barrierblood - brain barrier
blood - brain barrier
 
ChowetalTINSreview.pdf
ChowetalTINSreview.pdfChowetalTINSreview.pdf
ChowetalTINSreview.pdf
 
Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013Poster_Emerging Scholars Program 2013
Poster_Emerging Scholars Program 2013
 
Drug delivery across BBB
Drug delivery across BBBDrug delivery across BBB
Drug delivery across BBB
 
ijms-23-15271.pdf
ijms-23-15271.pdfijms-23-15271.pdf
ijms-23-15271.pdf
 
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf
5. The Blood-Brain Barrier. David S. Younger. Neurol Clinics 2019_37_235-248.pdf
 
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...
Akhtar and Breunig-2015-Frontiers in Cellular Neuroscience - Barriers to post...
 
ASN Neuro-2016-Nikolakopoulou-
ASN Neuro-2016-Nikolakopoulou-ASN Neuro-2016-Nikolakopoulou-
ASN Neuro-2016-Nikolakopoulou-
 
PIIS0166223620302861.pdf
PIIS0166223620302861.pdfPIIS0166223620302861.pdf
PIIS0166223620302861.pdf
 
Neuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into MemoriesNeuroscience: Transforming Visual Percepts into Memories
Neuroscience: Transforming Visual Percepts into Memories
 
Muthusamy_et_al-2015-Aging_Cell
Muthusamy_et_al-2015-Aging_CellMuthusamy_et_al-2015-Aging_Cell
Muthusamy_et_al-2015-Aging_Cell
 
C-CEBH__ Current Trainees_2015
C-CEBH__ Current Trainees_2015C-CEBH__ Current Trainees_2015
C-CEBH__ Current Trainees_2015
 
Jap 1 anat pathol
Jap 1 anat pathol Jap 1 anat pathol
Jap 1 anat pathol
 
Jnnt 3-016-1
Jnnt 3-016-1Jnnt 3-016-1
Jnnt 3-016-1
 
Project: defining the endothelial hematopoietic symbiosis
Project: defining the endothelial hematopoietic symbiosisProject: defining the endothelial hematopoietic symbiosis
Project: defining the endothelial hematopoietic symbiosis
 
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers Cns
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers CnsPharmacopsychiatry Bmsystems Max Planck Publication Biomarkers Cns
Pharmacopsychiatry Bmsystems Max Planck Publication Biomarkers Cns
 
Cell-Replacement Therapy with Stem Cells in Neurodegenerative Diseases
Cell-Replacement Therapy with Stem Cells in Neurodegenerative DiseasesCell-Replacement Therapy with Stem Cells in Neurodegenerative Diseases
Cell-Replacement Therapy with Stem Cells in Neurodegenerative Diseases
 
Research article the turing machine theory for some spinal cord and brain con...
Research article the turing machine theory for some spinal cord and brain con...Research article the turing machine theory for some spinal cord and brain con...
Research article the turing machine theory for some spinal cord and brain con...
 
B978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdfB978-0-444-63956-1.00002-3.pdf
B978-0-444-63956-1.00002-3.pdf
 
Blood brain barrier
Blood brain barrier Blood brain barrier
Blood brain barrier
 

Grab Chakravorty Heyde and STINS microbial interactions neuronal function 2011

  • 1. Microreview How can microbial interactions with the blood–brain barrier modulate astroglial and neuronal function? Dennis J. Grab,1 Srabasti J. Chakravorty,2 Henri van der Heyde3 and Monique F. Stins4 * 1 Johns Hopkins SOM, Department Pathology, Division Medical Microbiology, 720 Rutland Ave, Ross 624, Baltimore, MD 21205, USA. 2 Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK. 3 La Jolla Infectious Disease Institute, Bunker Hill Rd, La Jolla, CA, USA. 4 Johns Hopkins SOM, Department Neurology, NeuroSurgery and Psychiatry, Division NeuroImmunology, 600 N Wolfe street, Meyer 5-134 (mail 6-113), Baltimore, MD 21281, USA. Summary The vascular endothelium of the blood–brain barrier (BBB) is regarded as a part of the neurovas- cular unit (NVU). This emerging NVU concept emphasizes the need for homeostatic signalling among the neuronal, glial and vascular endothelial cellular compartments in maintaining normal brain function. Conversely, dysfunction in any compo- nent of the NVU affects another, thus contributing to disease. Brain endothelial activation and dys- function is observed in various neurological dis- eases, such as (ischemic) stroke, seizure, brain inflammation and infectious diseases and likely contributes to or exacerbates neurological condi- tions. The role and impact of brain endothelial factors on astroglial and neuronal activation is unclear. Similarly, it is not clear which stages of BBB endothelial activation can be considered beneficial versus detrimental. Although the BBB plays an important role in context of encephalopa- thies caused by neurotropic microbes that must first penetrate into the brain, a crucial role of the BBB in contributing to neurological dysfunction may be seen in cerebral malaria (CM), where the Plasmodium parasite remains sequestered in the brain vasculature, does not enter the brain paren- chyma, and yet causes coma and seizures. In this minireview some of the scenarios and factors that may play a role in BBB as a relay station to modu- late astroneuronal functioning are discussed.cmi_1661 1470..1478 The blood–brain barrier endothelium and the neurovascular unit The blood–brain barrier (BBB) endothelium forms a highly selective barrier between the blood and the central nervous system (CNS). Once regarded as ‘just’ a lining for vessels to allow blood to flow and nutrients to pass into the underlying tissues, it is now clear that endothelium in different tissues is very heterogeneous in its expression of surface receptors and responses to stimuli (Langenkamp and Molema, 2009). Contrary to liver endothelium that displays fenestrae to facilitate plasma exchange, brain endothelium possesses tight junctions characteristic of its barrier function. The unique tissue-specific endothelial characteristics are induced through interactions with the underlying tissues and in the CNS this includes the under- lying extracellular matrix, astrocytes and pericytes (Abbott et al., 2006; Kroll et al., 2009; Bonkowski et al., 2011; Kamouchi et al., 2011). In addition, brain-trophic BBB- derived factors can promote differentiation of neuropre- cursor cells (Guo et al., 2008; Chintawar et al., 2009). Indeed, the BBB endothelium (BBB-EC) is now regarded an integral component of the neurovascular unit (NVU), an emerging concept that underlines the importance of the homeostatic interactions between the brain’s cellular components – astroglia, neurons and BBB-EC – for optimal CNS functioning (Abbott et al., 2006; Persidsky et al., 2006; Lok et al., 2007; Bonkowski et al., 2011). Conversely, deregulation in any of the NVU components, will alter the brains homeostasis leading to a multitude of neurodysfunctions and BBB activation as has been observed in multiple sclerosis, Alzheimers disease, stroke, certain depression disorders and in microbial infections, among others (Zlokovic, 2008; Reale et al., 2009; Chung et al., 2010; Holman et al., 2011). The Received 21 June, 2011; revised 2 August, 2011; accepted 3 August, 2011. *For correspondence. E-mail mstins@jhmi.edu; Tel. (+1) 443 287 8027 Cellular Microbiology (2011) 13(10), 1470–1478 doi:10.1111/j.1462-5822.2011.01661.x First published online 6 September 2011 © 2011 Blackwell Publishing Ltd cellular microbiology
  • 2. contribution of several endothelial-activating components, including microbial involvement, has been suggested to initiate certain brain diseases (Miklossy, 2008; Zhang et al., 2009; Humpel, 2011; Ochoa-Reparaz et al., 2011). Circulating gut-derived lipopolysaccharides (LPS), which are increased in, e.g. HIV-1 infection, alcohol abuse and depression disorders, can activate brain endothelium to trigger astroglial activation signals and/or modulate neu- rological functioning that lead to sickness-related behav- ioural changes (Schafer et al., 2002; Maes et al., 2008). Modulation of microglial function by peripheral inflamma- tion also depends on the prior history and immune status (Cunningham et al., 2009). Unfortunately, the role of BBB-EC activation in the development of neurological diseases is still an understudied area of research. In vitro BBB endothelial models to study its involvement in neurological disease Single- or multi-cell in vitro models of the BBB have proven useful tools to study BBB-EC and its functionality in neurological diseases. For elucidation of specific BBB human endothelium responses, cultures comprising of only primary or immortalized human brain EC are pre- ferred (Stins et al., 2001a; Weksler et al., 2005). However, other models include non-human- and non-brain- endothelial cell types such as human umbilical vein endot- helial cells (Weiss et al., 1999; Eugenin et al., 2006), ECV304/T4 cells (Tan et al., 2001; Neuhaus et al., 2011), primary porcine brain endothelial cultures (Kroll et al., 2009), immortalized rat cultures (Roux and Couraud, 2005) and bovine brain endothelial cells (Stins et al., 1997). Today, a wide availability of diverse immortalized cell lines have become attractive workhorses and have greatly facilitated BBB research. However, because immortalized lines may have changed their physiology and do not always reflect the heterogeneity of endothelial populations and responses, it is imperative to verify results with primary brain endothelial cells that are now commercially available. With these tools, research to better understand the intimate interactions between BBB-EC and other cellular components within NVU and its contributions to neurological disease is now universally possible. To elicit more BBB-like characteristics, BBB-EC can be cultured on brain-specific extracellular matrix, in combina- tion multi-cell co-cultures, either with pericytes, astro- cytes, C-6 glioma cells or in the presence of conditioned media from these cells (Nakagawa et al., 2009; Hatherell et al., 2011). The ‘modular’ system approach allows studies on direct interactions between the different cell types of the NVU, which may not be possible in vivo. However, astrocytes are heterogeneous and can be ‘acti- vated’ upon isolation (Zhang and Barres, 2010), either positively or negatively impacting on the BBB-EC charac- teristics when used in co-culture models. Physical factors, such as vascular sheer stress and red blood cell (RBC) interactions, can affect BBB characteristics such as higher trans endothelial electrical resistance (TEER), decreased permeability and altered expression of multi- drug transporters (Tripathi et al., 2007; Cucullo et al., 2011a,b). Complex hollow tube dynamic flow models and two-dimensional flow models allow for studies on interactions of (microbial infected-) immune cells and/or RBCs under flow (Barth et al., 2009; Phiri et al., 2009) with BBB-EC. Microbes, BBB and neurological dysfunction; to cross or not to cross? Generally, high plasma concentrations of neurotropic microbes (e.g. sepsis) are a prerequisite for CNS entry. BBB crossing can occur via transcellular or paracellular mechanisms, or when immune cells are ‘hijacked’ by microbes in the periphery, by Trojan Horse mechanisms (Toborek et al., 2005; Pulzova et al., 2009; Elsheikha and Khan, 2010; Kaushik et al., 2011). Once inside the CNS, the neurons, microglia or astrocytes can be infected or activated by microbial toxins, such as LPS. Innate immu- nity plays an important role here, as the microbial ligands are sensed by toll-like receptors (TLRs) leading to signal transduction via NFkB. The subsequent release of (inflammatory) mediators by the activated astroglia might then alter neurological function and affect BBB function and integrity (Kang and McGavern, 2010). Remarkably, not all pathogens that cause neurological disease breach the vascular barrier and enter the CNS. In low numbers, circulating microbes and their toxins might not penetrate into the CNS but can still activate the BBB-EC to release signals that affect the underlying neuropil (i.e. brain cells) (Fig. 2). Mild stimulation of the brain endothelium with low amounts of microbes/toxins increases expression of cell adhesion molecules and release of cytokines/chemokines, whereas stronger stimulation also includes opening of the intercellular junc- tions and increased barrier permeability (Tripathi et al., 2006; 2009), thus exposing the brain to neurotoxic plasma substances, including serum albumin that can induce seizures (Friedman et al., 2009; Hooper et al., 2009) or antibodies to ion channels, thus affecting neurological function (Lang et al., 2005). Little is known how brain EC activation by itself can contribute to neuropathogenesis. Here, we examine microbial CNS infections in the context of the BBB as a relay station and modulator of astroglial– neuronal function. A fascinating example of peripheral influences that affect neurological functioning can be appreciated in cere- bral malaria (CM) where the Plasmodium falciparum para- How can the blood–brain barrier endothelium influence the brain? 1471 © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 3. site that lives inside an infected RBC (PRBC) ‘remodels’ the PRBC surface with parasite-encoded proteins (Pf- EMP-1) that then bind to endothelial receptors such as ICAM-1, resulting in a sequestration of PRBCs in the vascular lumen. PRBC sequestration is considered central to CM pathology and concomitant with other brain- endothelial activating factors likely contributes to the neu- rological symptoms (Turner et al., 1994; Adams et al., 2002; van der Heyde et al., 2006; Combes et al., 2010). Non-immune adults (e.g. travellers) and mostly young children are especially susceptible to CM, manifested by impaired consciousness, seizures and coma (Idro et al., 2005). Unfortunately, even after successful treatment and recovery, neurological sequelae can remain (Idro et al., 2010). Understanding CM pathogenesis is therefore of utmost importance and may clarify the BBB role in other neurological diseases. In our laboratories we are currently studying the mechanisms whereby PRBC induce neurological dysfunction. Using Transwelltm -based in vitro human BBB models validated for CM research (Tripathi et al., 2006; 2007; 2009), we found that PRBC- mediated activation of the host BBB-EC leads to increase expression of luminal ICAM-1 and polarized release of cytokines/chemokine to both luminal (blood-facing) side and basal (brain-facing) side of the monolayers. A signifi- cant loss of barrier integrity was only observed upon addi- tion of high numbers of PRBC. Traditionally cytokines/ chemokines are thought to be released to the luminal side of the BBB in order to alarm the immune system. However, circulating cytokines can also shuttle across the BBB into the CNS (Banks et al., 1995) and both luminal and basal release of cytokines from BBB-ECs in response to LPS has also been described (Verma et al., 2006). To test if the BBB secretions towards the brain side would affect astrocytes and/or neurons, we collected this con- ditioned BBB medium from our CM experiments and added it to astrocyte and neuronal cultures (Fig. 1). Here, we found a concentration-dependent activation of astro- cytes (by GFAP immunocytochemistry) and in neurons disruption of axonal transport and retraction of neuronal extensions (Stins laboratory, unpubl. obs.). This is in agreement with axonal damage as found in human CM autopsies (Medana et al., 2007). These cytokines/ chemokines present in the basal BBB-EC secretions may play a role in glial cell migration, angiogenesis, tumorigen- esis, and wound healing, but also modulate astrocyte– neuronal interactions and thus be involved in neurological dysfunction. BBB activation by other pathogens In advanced stages of Toxoplasma gondii encephalitis (TE), BALB/c and more sensitive C57BL/6 murine models show BBB activation and damage as reflected by (i) a prominent induction of major histocompatibility complex (MHC) class I and II antigens, (ii) cell adhesion molecules VCAM-1 and ICAM-1 expression (Deckert-Schluter et al., 1999; Wang et al., 2007), (iii) a higher ALCAM expression and (iv) increased BBB permeability (Silva et al., 2010). T. gondii binds and infects retinal-brain endothelial cells, bovine umbilical vein endothelial cells (BUVEC) and human endothelium in vitro, leading to BBB activation. In retinal EC and BUVEC, T. gondii induces increases in transcripts for E-, P-selectin, VCAM-1 and ICAM-1 within 1–4 h and inflammatory mediators including GRO1, MCP-1, Fractalkine, RANTES, IL8, IP10, MCP-1, GM-CSF, COX2 and iNOS persist at least up to 72 h post infection (Daubener et al., 2001; Stumbo et al., 2002; Fig. 1. In vitro set-up to study effects of brain endothelium on underlying brain cells. Shown here is a Transwelltm tissue culture insert that can be seeded with brain endothelial cells and inserted in a tissue culture wells, thus creating a two-chamber model where the upper compartment represents the blood side and the bottom compartment the brain side. Dependent on the experimental set-up astrocytes, pericytes can be incorporated on the abluminal side on the membrane (in contact with the brain endothelial cells) (B), in the bottom compartment or in a separate tissue culture well (A). Upon confluence and when sufficient barrier function is obtained, microbes can be added to the upper compartment. Activation of the BBB endothelium, release of factors to either side and their effects on the underlying brain cells can then be studied. 1472 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 4. Smith et al., 2004; Knight et al., 2005; Taubert et al., 2006a,b). Once T. gondii crosses the BBB, subsequent microglial infection and astrocyte activation can then have a negative effect on the BBB leading to activation and loss of integrity (Silva et al., 2010; Dellacasa-Lindberg et al., 2011). African trypanosomes (Trypanosoma brucei spp.) upregulate brain endothelial expression of ICAM-1, VCAM-1 and E-selectin and release of IL-6, CXCL8, CCL2 and TNF-a (Mulenga et al., 2001; Girard et al., 2005; Grab and Kennedy, 2008). Matrix metalloprotease MMP2 was increased in CSF (Hainard et al., 2011) and in vitro studies showed that brain endothelium can poten- tially contribute to this (D. Grab, unpublished). In vitro and gene-profiling studies showed that Trypanosome transmi- gration requires cysteine proteases Gaq-mediated calcium and protease-activated receptor-2 (PAR-2) sig- nalling (Nikolskaia et al., 2006; Grab et al., 2009a), initial events that are precursory to CNS neuroinflammatory disease (Grab et al., 2009a). Activation of BBB-EC by Borreliia burgdorferi alone or in co-infection with Anaplasma phygocytophilum- infected neutrophils has also been observed. Increased expression of host cell adhesion molecules, plasmino- gen activators, plasminogen activator receptors and increases release of cytokines, chemokines and metal- loproteases might contribute to neuropathogenesis (Sellati et al., 1995; Burns et al., 1997; Grab et al., 2005; 2007; 2009b). Blood–brain barrier breakdown as a loss of junctional molecules has been shown during advanced neuro-HIV infection based on biopsy and post-mortem samples and in vivo disease models (Persidsky et al., 2006; van Horssen et al., 2007). During initial infection, high viral titres and secreted viral proteins activate the BBB-EC resulting in increases in expression of cell adhesion mol- ecules and release of cytokines and chemokines (Stins et al., 2001b; 2004; Persidsky et al., 2006; Shiu et al., 2007; Chaudhuri et al., 2008), which directed towards the brain side can affect neurological functioning and contrib- utes to sickness behaviour. The dysregulation of the BBB during and after neuroinvasion is a critical component of the neuropathogenic process (Strazza et al., 2011). An indirect pathogen-induced peripheral stimulus can also alter neurological function and behaviour. Circulating LPS, which is present in low concentrations in conditions including HIV infection, alcohol abuse and some major depression disorders (Frank et al., 2004; Maes et al., 2008; Nowroozalizadeh et al., 2010), does not readily enter the CNS, but accumulates in the BBB-EC, but still astroglial activation is observed (Singh and Jiang, 2004; Banks and Robinson, 2009). Mechanisms for BBB-related neurological modulation In neurotropic infections, generally similar host responses are observed at the BBB, but a role of the BBB in confer- ring neurological dysfunction is most clearly observed in CM. However, the mechanisms that are responsible are unclear, as is their exact function and how they will affect neuronal function. Reduced release of neuroprotective Fig. 2. Transmission of astroglial and neuronal modulating signals from the BBB endothelium. Neurotropic microbes, like PRBC, HIV-1 or Trypanosomes, can stimulate the release of soluble factors (represented by the arrows), such as astroglial-activating matrix metalloproteases, cytokines and chemokines such as IL-1b, IL-6, CCL20, GRO’s and TNFa from the basal side of brain endothelial cells. Targets for these mediators are present in the brain on either astroglial cells and/or neurons and can result in modulation of astroglial activation and both directly and indirectly of neuronal function. Vessel lumen Microbial ac!va!on = Cell adhesion molecules = signaling BBB- endothelium Brain parenchyma endothelium NO + ? MMP’s Cytokines Chemokines Astrocytes/ microglia + -? + -? Neuron + -? Neurological Modula!on > BeneÞts versus Complica!ons How can the blood–brain barrier endothelium influence the brain? 1473 © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 5. secretions like brain-derived neurotrophic factor (BDNF) versus increases in release of nitric oxide (NO), glycine, cytokines, chemokines and/or MMPs could contribute to these effects (Fig. 2). Dependent on the individual, release of BBB-EC-derived cytokines/chemokines could be very robust in CM (Tripathi et al., 2009; Wassmer et al., 2011). Inside the CNS, the BBB-derived cytokines/ chemokines may play a role in glial cell migration, angio- genesis, tumorigenesis and wound healing, but may also modulate astrocyte–neuronal interactions and thus be involved in neurological dysfunction. At this moment, it is unclear as to which particular cytokines/chemokines are involved, their exact function, how they will affect neuronal functioning and whether their targets are on brain cells. Using in vitro BBB-CM models we observed large increases in CCL20 transcripts and release (Tripathi et al., 2009). The non-promiscuous receptor for CCL20, CCR6 is generally non-detectable in control brains, but upregulated in brain inflammation, such as in the murine model for CM that replicates certain features of human pathology (Hunt et al., 2010), and we see increased CCR6 immunostaining in cells resembling neuroglia, possible oligodendrocytes. Oligodendrocytes are believed to be involved in neuronal signal modula- tion, and as such, BBB-released CCL20 could affect neuronal transmission. Astrocytes also secrete CXCL1, 2, 3 and their receptor (CXCR2) is present on neuron (Dorf et al., 2000). CXCR2 can regulate APMA type glutamate receptor function by increasing receptor coop- erativety and postsynaptic transmission amplitude (Lax et al., 2002). Furthermore, IL8 and CXCL1 can modulate Purkinje neuron activity (Giovannelli et al., 1998). CXCL1 increases ERK phosphorylation in cortical neurons (Xia and Hyman, 2002). Thus, these may be potential mechanisms whereby release of CXCL1, 2 or 3 from the basolateral side of the BBB, may modulate astroglial and/or neuronal function. Taken together, alter- ation of the chemokine/receptor ratio in the brain may be a mechanism to modulate neuronal function, but when the BBB is over activated, this may lead to neuronal dysfunction. Other factors, such as MMPs or NO basally released from an activated BBB can modulate not only matrix– BBB-EC interactions and thus barrier integrity but also synaptic function and long-term potentiation by cleavage of NR1 subunit of the NMDA receptor and ICAM-5 (Szklarczyk et al., 2002; Huang et al., 2008; Conant et al., 2010; Louboutin et al., 2010). In CM, NO released from the BBB can have a dual effect; luminal released NO is quenched by free haemoglobin and reactive oxygen species and therefore results in a low peripheral NO bioavailability (Gramaglia, 2006). As serum arginine levels are coupled to eNOS activity, it is not clear how this affects NO released to the basal side of the BBB in CM and other neuromicrobial diseases. NO has various effects on neuronal function, including anterograde signalling (Fernandez-Alvarez et al., 2011), cognitive learning (Paul and Ekambaram, 2011), depres- sion behaviour (Workman et al., 2011), glutamatergic transmission (Sardo et al., 2011) and altering of sensi- tivity of dopamine release to presynaptic activation (Hartung et al., 2011). In astrocytes, NO alters connex- ins leading to reduction of inter-astrocytic gap-junction communications (Ball et al., 2011) which could also affect astrocyte–BBB-EC or astrocyte–neurons com- munications. Reactive nitrogen species can trigger lipid peroxidation chain reactions throughout the cerebral vasculature, and brain parenchyma and thus contribut- ing neurocognitive sequelae as seen in sepsis survivors (Berg et al., 2011). Conclusions It makes sense that, in the context of the NVU principle, an alteration in the ‘resting’ status of the BBB-EC by neurotropic microbes can lead to modulation of astroglial/neuronal function. Thus, an activated BBB could function as a communication-relay station between peripheral signals to the brain and contribute to symp- toms ranging from mild symptoms such as malaise to serious neurological dysfunction such as coma in CM. Brain endothelial-derived modulating factors that play a role in NVU function include the and increased release of local and brain-directed cytokines, chemokines, NO and MMPs versus a decreased release of neurotrophic and protective factors. It is not clear at what point the altered release of these factors would tip the balance from playing a beneficial role in astroneuronal homeo- stasis and repair to a detrimental one leading to neuro- logical dysfunction (Fig. 3). Therefore, it is important to understand BBB-EC factors that are involved in mediat- ing and regulating the responses to microbial exposure and ‘decide’ at which point the balance tips from a ben- eficial neuroprotective response to a detrimental reac- tion. BBB responses, like in CM or stroke, can be very local and affecting only small brain areas. Therefore, changes in these factors can be missed in systemic measurements of blood or CSF samples. In vitro systems can aid to identify these factors that subse- quently can be validated in vivo. Many questions still remain and further clarification of interactions between the BBB-EC and underlying CNS components are needed as this may not only lead to novel therapeutic targets for treating neurological dysfunction in a wide variety of disorders but could also be beneficial in the repair of astroneuronal function in conditions such as stroke. 1474 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 6. References Abbott, N.J., Ronnback, L., and Hansson, E. (2006) Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci 7: 41–53. Adams, S., Brown, H., and Turner, G. (2002) Breaking down the blood–brain barrier: signaling a path to cerebral malaria? Trends Parasitol 18: 360–366. Ball, K.K., Harik, L., Gandhi, G.K., Cruz, N.F., and Dienel, G.A. (2011) Reduced gap junctional communication among astrocytes in experimental diabetes: contributions of altered connexin protein levels and oxidative-nitrosative modifications. J Neurosci Res doi: 10.1002/jnr.22663 Banks, W.A., and Robinson, S.M. (2009) Minimal penetration of lipopolysaccharide across the murine blood–brain barrier. Brain Behav Immun 24: 102–109. Banks, W.A., Kastin, A.J., and Broadwell, R.D. (1995) Passage of cytokines across the blood–brain barrier. Neu- roimmunomodulation 2: 241–248. Barth, M.C., Ahluwalia, N., Anderson, T.J., Hardy, G.J., Sinha, S., Alvarez-Cardona, J.A., et al. (2009) Kynurenic acid triggers firm arrest of leukocytes to vascular endothe- lium under flow conditions. J Biol Chem 284: 19189– 19195. Berg, R.M., Moller, K., and Bailey, D.M. (2011) Neuro- oxidative-nitrosative stress in sepsis. J Cereb Blood Flow Metab 31: 1532–1544. Bonkowski, D., Katyshev, V., Balabanov, R.D., Borisov, A., and Dore-Duffy, P. (2011) The CNS microvascular pericyte: pericyte-astrocyte crosstalk in the regulation of tissue sur- vival. Fluids Barriers CNS 8: 8. Burns, M.J., Sellati, T.J., Teng, E.I., and Furie, M.B. (1997) Production of interleukin-8 (IL-8) by cultured endothelial cells in response to Borrelia burgdorferi occurs indepen- dently of secreted [corrected] IL-1 and tumor necrosis factor alpha and is required for subsequent transendothe- lial migration of neutrophils. Infect Immun 65: 1217–1222. Chaudhuri, A., Duan, F., Morsey, B., Persidsky, Y., and Kanmogne, G.D. (2008) HIV-1 activates proinflammatory and interferon-inducible genes in human brain microvascu- lar endothelial cells: putative mechanisms of blood–brain barrier dysfunction. J Cereb Blood Flow Metab 28: 697– 711. Chintawar, S., Cayrol, R., Antel, J., Pandolfo, M., and Prat, A. (2009) Blood–brain barrier promotes differentiation of human fetal neural precursor cells. Stem Cells 27: 838–846. Chung, Y.C., Ko, H.W., Bok, E., Park, E.S., Huh, S.H., Nam, J.H., et al. (2010) The role of neuroinflammation on the pathogenesis of Parkinson’s disease. BMB Rep 43: 225– 232. Combes, V., El-Assaad, F., Faille, D., Jambou, R., Hunt, N.H., and Grau, G.E. (2010) Microvesiculation and cell interac- tions at the brain–endothelial interface in cerebral malaria pathogenesis. Prog Neurobiol 91: 140–151. Conant, K., Wang, Y., Szklarczyk, A., Dudak, A., Mattson, M.P., and Lim, S.T. (2010) Matrix metalloproteinase- dependent shedding of intercellular adhesion molecule-5 occurs with long-term potentiation. Neuroscience 166: 508–521. Cucullo, L., Hossain, M., Puvenna, V., Marchi, N., and Janigro, D. (2011a) The role of shear stress in Blood–Brain Barrier endothelial physiology. BMC Neurosci 12: 40. Cucullo, L., Marchi, N., Hossain, M., and Janigro, D. (2011b) A dynamic in vitro BBB model for the study of immune cell trafficking into the central nervous system. J Cereb Blood Flow Metab 31: 767–777. Cunningham, C., Campion, S., Lunnon, K., Murray, C.L., Woods, J.F., Deacon, R.M., et al. (2009) Systemic inflam- mation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychia- try 65: 304–312. Daubener, W., Spors, B., Hucke, C., Adam, R., Stins, M., Kim, K.S., et al. (2001) Restriction of Toxoplasma gondii growth in human brain microvascular endothelial cells by activation of indoleamine 2,3-dioxygenase. Infect Immun 69: 6527–6531. Deckert-Schluter, M., Bluethmann, H., Kaefer, N., Rang, A., and Schluter, D. (1999) Interferon-gamma receptor- mediated but not tumor necrosis factor receptor type 1- or type 2-mediated signaling is crucial for the activation of Fig. 3. The balance of factors derived from the activated BBB endothelium could determine a neuroprotective versus detrimental response. Under homeostatic conditions, the BBB endothelium releases substances such as cytokines, chemokines, NO and MMPs, neurotrophic and neuroprotective factors towards the brain side. Upon activation, the BBB endothelium releases factors in order to ‘alert’ brain cells and protect the neurological function. However, with strong activation the BBB may ‘overreact’ and these otherwise positive factors may turn into detrimental substances. A decrease in neuroprotective and neurotrophic secretions may occur concomitantly. It is not yet clear which BBB-EC factors govern the release of these substances and at what point the balance tips from being protective of the brain’s homeostasis to being negative and affecting astroneuronal function. How can the blood–brain barrier endothelium influence the brain? 1475 © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 7. cerebral blood vessel endothelial cells and microglia in murine Toxoplasma encephalitis. Am J Pathol 154: 1549– 1561. Dellacasa-Lindberg, I., Fuks, J.M., Arrighi, R.B., Lambert, H., Wallin, R.P., Chambers, B.J., et al. (2011) Migratory acti- vation of primary cortical microglia upon infection with Toxoplasma gondii. Infect Immun 79: 3046–3052. Dorf, M.E., Berman, M.A., Tanabe, S., Heesen, M., and Luo, Y. (2000) Astrocytes express functional chemokine recep- tors. J Neuroimmunol 111: 109–121. Elsheikha, H.M., and Khan, N.A. (2010) Protozoa traversal of the blood–brain barrier to invade the central nervous system. FEMS Microbiol Rev 34: 532–553. Eugenin, E.A., Osiecki, K., Lopez, L., Goldstein, H., Calderon, T.M., and Berman, J.W. (2006) CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigra- tion of human immunodeficiency virus (HIV)-infected leukocytes across the blood–brain barrier: a potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci 26: 1098–1106. Fernandez-Alvarez, A., Gomez-Sena, L., Fabbiani, M.G., Budelli, R., and Abudara, V. (2011) Endogenous pre- synaptic nitric oxide supports an anterograde signaling in the central nervous system. J Neurochem 118: 546– 557. Frank, J., Witte, K., Schrodl, W., and Schutt, C. (2004) Chronic alcoholism causes deleterious conditioning of innate immunity. Alcohol Alcohol 39: 386–392. Friedman, A., Kaufer, D., and Heinemann, U. (2009) Blood– brain barrier breakdown-inducing astrocytic transforma- tion: novel targets for the prevention of epilepsy. Epilepsy Res 85: 142–149. Giovannelli, A., Limatola, C., Ragozzino, D., Mileo, A.M., Ruggieri, A., Ciotti, M.T., et al. (1998) CXC chemokines interleukin-8 (IL-8) and growth-related gene product alpha (GROalpha) modulate Purkinje neuron activity in mouse cerebellum. J Neuroimmunol 92: 122–132. Girard, M., Giraud, S., Courtioux, B., Jauberteau-Marchan, M.O., and Bouteille, B. (2005) Endothelial cell activation in the presence of African trypanosomes. Mol Biochem Para- sitol 139: 41–49. Grab, D.J., and Kennedy, P.G. (2008) Traversal of human and animal trypanosomes across the blood–brain barrier. J Neurovirol 14: 344–351. Grab, D.J., Perides, G., Dumler, J.S., Kim, K.J., Park, J., Kim, Y.V., et al. (2005) Borrelia burgdorferi, host-derived pro- teases, and the blood–brain barrier. Infect Immun 73: 1014–1022. Grab, D.J., Nyarko, E., Barat, N.C., Nikolskaia, O.V., and Dumler, J.S. (2007) Anaplasma phagocytophilum–Borrelia burgdorferi coinfection enhances chemokine, cytokine, and matrix metalloprotease expression by human brain microvascular endothelial cells. Clin Vaccine Immunol 14: 1420–1424. Grab, D.J., Garcia-Garcia, J.C., Nikolskaia, O.V., Kim, Y.V., Brown, A., Pardo, C.A., et al. (2009a) Protease activated receptor signaling is required for African trypanosome tra- versal of human brain microvascular endothelial cells. PLoS Negl Trop Dis 3: e479. Grab, D.J., Nyarko, E., Nikolskaia, O.V., Kim, Y.V., and Dumler, J.S. (2009b) Human brain microvascular endo- thelial cell traversal by Borrelia burgdorferi requires calcium signaling. Clin Microbiol Infect 15: 422–426. Gramaglia, I. Low nitric oxide bioavailability contributes to the genesis of experimental cerebral malaria. Sobolewski, P., Meays, D., Contreras, R., Nolan, J.P., Frangos, J.A., Inta- glietta, M., and van der Heyde, H.C. Nat Med 12 [12], 1417–1422. 2006. Ref Type: Generic. Guo, S., Kim, W.J., Lok, J., Lee, S.R., Besancon, E., Luo, B.H., et al. (2008) Neuroprotection via matrix-trophic cou- pling between cerebral endothelial cells and neurons. Proc Natl Acad Sci USA 105: 7582–7587. Hainard, A., Tiberti, N., Robin, X., Ngoyi, D.M., Matovu, E., Enyaru, J.C., et al. (2011) Matrix metalloproteinase-9 and intercellular adhesion molecule 1 are powerful staging markers for human African trypanosomiasis. Trop Med Int Health 16: 119–126. Hartung, H., Threlfell, S., and Cragg, S.J. (2011) Nitric oxide donors enhance the frequency dependence of dopamine release in nucleus accumbens. Neuropsychopharmacol- ogy 36: 1811–1822. Hatherell, K., Couraud, P.O., Romero, I.A., Weksler, B., and Pilkington, G.J. (2011) Development of a three- dimensional, all-human in vitro model of the blood–brain barrier using mono-, co-, and tri-cultivation Transwell models. J Neurosci Methods 199: 223–229. van der Heyde, H.C., Nolan, J., Combes, V., Gramaglia, I., and Grau, G.E. (2006) A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemo- stasis leading to microcirculatory dysfunction. Trends Para- sitol 22: 503–508. Holman, D.W., Klein, R.S., and Ransohoff, R.M. (2011) The blood–brain barrier, chemokines and multiple sclerosis. Biochim Biophys Acta 1812: 220–230. Hooper, C., Pinteaux-Jones, F., Fry, V.A., Sevastou, I.G., Baker, D., Heales, S.J., et al. (2009) Differential effects of albumin on microglia and macrophages; implications for neurodegeneration following blood–brain barrier damage. J Neurochem 109: 694–705. van Horssen, J., Brink, B.P., de Vries, H.E., van der Valk, P., and Bo, L. (2007) The blood–brain barrier in cortical mul- tiple sclerosis lesions. J Neuropathol Exp Neurol 66: 321– 328. Huang, W., Rha, G.B., Han, M.J., Eum, S.Y., Andras, I.E., Zhong, Y., et al. (2008) PPARalpha and PPARgamma effectively protect against HIV-induced inflammatory responses in brain endothelial cells. J Neurochem 107: 497–509. Humpel, C. (2011) Chronic mild cerebrovascular dysfunction as a cause for Alzheimer’s disease? Exp Gerontol 46: 225–232. Hunt, N.H., Grau, G.E., Engwerda, C., Barnum, S.R., van der Heyde, H., Hansen, D.S., et al. (2010) Murine cerebral malaria: the whole story. Trends Parasitol 26: 272– 274. Idro, R., Jenkins, N.E., and Newton, C.R. (2005) Pathogen- esis, clinical features, and neurological outcome of cere- bral malaria. Lancet Neurol 4: 827–840. Idro, R., Kakooza-Mwesige, A., Balyejjussa, S., Mirembe, G., Mugasha, C., Tugumisirize, J., et al. (2010) Severe neuro- logical sequelae and behaviour problems after cerebral malaria in Ugandan children. BMC Res Notes 3: 104. 1476 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 8. Kamouchi, M., Ago, T., and Kitazono, T. (2011) Brain peri- cytes: emerging concepts and functional roles in brain homeostasis. Cell Mol Neurobiol 31: 175–193. Kang, S.S., and McGavern, D.B. (2010) Microbial induction of vascular pathology in the CNS. J Neuroimmune Pharmacol 5: 370–386. Kaushik, D.K., Gupta, M., and Basu, A. (2011) Microglial response to viral challenges: every silver lining comes with a cloud. Front Biosci 17: 2187–2205. Knight, B.C., Brunton, C.L., Modi, N.C., Wallace, G.R., and Stanford, M.R. (2005) The effect of Toxoplasma gondii infection on expression of chemokines by rat retinal vas- cular endothelial cells. J Neuroimmunol 160: 41–47. Kroll, S., El-Gindi, J., Thanabalasundaram, G., Panpumthong, P., Schrot, S., Hartmann, C., et al. (2009) Control of the blood–brain barrier by glucocorticoids and the cells of the neurovascular unit. Ann N Y Acad Sci 1165: 228– 239. Lang, B., Newbold, C.I., Williams, G., Peshu, N., Marsh, K., and Newton, C.R. (2005) Antibodies to voltage-gated calcium channels in children with falciparum malaria. J Infect Dis 191: 117–121. Langenkamp, E., and Molema, G. (2009) Microvascular endothelial cell heterogeneity: general concepts and phar- macological consequences for anti-angiogenic therapy of cancer. Cell Tissue Res 335: 205–222. Lax, P., Limatola, C., Fucile, S., Trettel, F., Di Bartolomeo, S., Renzi, M., et al. (2002) Chemokine receptor CXCR2 regu- lates the functional properties of AMPA-type glutamate receptor GluR1 in HEK cells. J Neuroimmunol 129: 66–73. Lok, J., Gupta, P., Guo, S., Kim, W.J., Whalen, M.J., van Leyen, K., et al. (2007) Cell–cell signaling in the neurovas- cular unit. Neurochem Res 32: 2032–2045. Louboutin, J.P., Agrawal, L., Reyes, B.A., Van Bockstaele, E.J., and Strayer, D.S. (2010) HIV-1 gp120-induced injury to the blood–brain barrier: role of metalloproteinases 2 and 9 and relationship to oxidative stress. J Neuropathol Exp Neurol 69: 801–816. Maes, M., Kubera, M., and Leunis, J.C. (2008) The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol Lett 29: 117–124. Medana, I.M., Day, N.P., Hien, T.T., Mai, N.T., Bethell, D., Phu, N.H., et al. (2007) Cerebral calpain in fatal falciparum malaria. Neuropathol Appl Neurobiol 33: 179–192. Miklossy, J. (2008) Chronic inflammation and amyloidogen- esis in Alzheimer’s disease – role of Spirochetes. J Alzheimers Dis 13: 381–391. Mulenga, C., Mhlanga, J.D., Kristensson, K., and Robertson, B. (2001) Trypanosoma brucei brucei crosses the blood– brain barrier while tight junction proteins are preserved in a rat chronic disease model. Neuropathol Appl Neurobiol 27: 77–85. Nakagawa, S., Deli, M.A., Kawaguchi, H., Shimizudani, T., Shimono, T., Kittel, A., et al. (2009) A new blood–brain barrier model using primary rat brain endothelial cells, peri- cytes and astrocytes. Neurochem Int 54: 253–263. Neuhaus, W., Freidl, M., Szkokan, P., Berger, M., Wirth, M., Winkler, J., et al. (2011) Effects of NMDA receptor modu- lators on a blood–brain barrier in vitro model. Brain Res 1394: 49–61. Nikolskaia, O.V., de A Lima, A.P., Kim, Y.V., Lonsdale-Eccles, J.D., Fukuma, T., Scharfstein, J., et al. (2006) Blood–brain barrier traversal by African trypanosomes requires calcium signaling induced by parasite cysteine protease. J Clin Invest 116: 2739–2747. Nowroozalizadeh, S., Mansson, F., da, S.Z., Repits, J., Dabo, B., Pereira, C., et al. (2010) Microbial translocation corre- lates with the severity of both HIV-1 and HIV-2 infections. J Infect Dis 201: 1150–1154. Ochoa-Reparaz, J., Mielcarz, D.W., Begum-Haque, S., and Kasper, L.H. (2011) Gut, bugs, and brain: role of commen- sal bacteria in the control of central nervous system disease. Ann Neurol 69: 240–247. Paul, V., and Ekambaram, P. (2011) Involvement of nitric oxide in learning & memory processes. Indian J Med Res 133: 471–478. Persidsky, Y., Ramirez, S.H., Haorah, J., and Kanmogne, G.D. (2006) Blood–brain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol 1: 223–236. Phiri, H., Montgomery, J., Molyneux, M., and Craig, A. (2009) Competitive endothelial adhesion between Plasmodium falciparum isolates under physiological flow conditions. Malar J 8: 214. Pulzova, L., Bhide, M.R., and Andrej, K. (2009) Pathogen translocation across the blood–brain barrier. FEMS Immunol Med Microbiol 57: 203–213. Reale, M., Iarlori, C., Thomas, A., Gambi, D., Perfetti, B., Di Nicola, M., et al. (2009) Peripheral cytokines profile in Par- kinson’s disease. Brain Behav Immun 23: 55–63. Roux, F., and Couraud, P.O. (2005) Rat brain endothelial cell lines for the study of blood–brain barrier permeability and transport functions. Cell Mol Neurobiol 25: 41–58. Sardo, P., Carletti, F., Rizzo, V., Lonobile, G., Friscia, S., and Ferraro, G. (2011) Nitric oxide-active compounds modulate the intensity of glutamate-evoked responses in the globus pallidus of the rat. Life Sci 88: 1113–1120. Schafer, C., Parlesak, A., Schutt, C., Bode, J.C., and Bode, C. (2002) Concentrations of lipopolysaccharide-binding protein, bactericidal/permeability-increasing protein, soluble CD14 and plasma lipids in relation to endotox- aemia in patients with alcoholic liver disease. Alcohol Alcohol 37: 81–86. Sellati, T.J., Burns, M.J., Ficazzola, M.A., and Furie, M.B. (1995) Borrelia burgdorferi upregulates expression of adhesion molecules on endothelial cells and promotes transendothelial migration of neutrophils in vitro. Infect Immun 63: 4439–4447. Shiu, C., Barbier, E., Di, C.F., Choi, H.J., and Stins, M. (2007) HIV-1 gp120 as well as alcohol affect blood–brain barrier permeability and stress fiber formation: involvement of reactive oxygen species. Alcohol Clin Exp Res 31: 130– 137. Silva, N.M., Manzan, R.M., Carneiro, W.P., Milanezi, C.M., Silva, J.S., Ferro, E.A., et al. (2010) Toxoplasma gondii: the severity of toxoplasmic encephalitis in C57BL/6 mice is associated with increased ALCAM and VCAM-1 expres- sion in the central nervous system and higher blood–brain barrier permeability. Exp Parasitol 126: 167–177. How can the blood–brain barrier endothelium influence the brain? 1477 © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478
  • 9. Singh, A.K., and Jiang, Y. (2004) How does peripheral lipopolysaccharide induce gene expression in the brain of rats? Toxicology 201: 197–207. Smith, J.R., Franc, D.T., Carter, N.S., Zamora, D., Planck, S.R., and Rosenbaum, J.T. (2004) Susceptibility of retinal vascular endothelium to infection with Toxoplasma gondii tachyzoites. Invest Ophthalmol Vis Sci 45: 1157–1161. Stins, M.F., Prasadarao, N.V., Zhou, J., Arditi, M., and Kim, K.S. (1997) Bovine brain microvascular endothelial cells transfected with SV40-large T antigen: development of an immortalized cell line to study pathophysiology of CNS disease. In Vitro Cell Dev Biol Anim 33: 243–247. Stins, M.F., Badger, J., and Sik Kim, K. (2001a) Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog 30: 19–28. Stins, M.F., Shen, Y., Huang, S.H., Gilles, F., Kalra, V.K., and Kim, K.S. (2001b) Gp120 activates children’s brain endot- helial cells via CD4. J Neurovirol 7: 125–134. Stins, M.F., Pearce, D., Choi, H., Di Cello, F., Pardo, C.A., and Kim, K.S. (2004) CD4 and chemokine receptors on human brain microvascular endothelial cells, implications for human immunodeficiency virus type 1 pathogenesis. Endothelium 11: 275–284. Strazza, M., Pirrone, V., Wigdahl, B., and Nonnemacher, M.R. (2011) Breaking down the barrier: the effects of HIV-1 on the blood–brain barrier. Brain Res 5: 1396–1399. Stumbo, A.C., Barbosa, H.S., Carvalho, T.M., Porto, L.C., and Carvalho, L. (2002) Anionic sites, fucose residues and class I human leukocyte antigen fate during interaction of Toxoplasma gondii with endothelial cells. Mem Inst Oswaldo Cruz 97: 517–522. Szklarczyk, A., Lapinska, J., Rylski, M., McKay, R.D., and Kaczmarek, L. (2002) Matrix metalloproteinase-9 under- goes expression and activation during dendritic remodeling in adult hippocampus. J Neurosci 22: 920–930. Tan, K.H., Dobbie, M.S., Felix, R.A., Barrand, M.A., and Hurst, R.D. (2001) A comparison of the induction of immor- talized endothelial cell impermeability by astrocytes. Neu- roreport 12: 1329–1334. Taubert, A., Krull, M., Zahner, H., and Hermosilla, C. (2006a) Toxoplasma gondii and Neospora caninum infections of bovine endothelial cells induce endothelial adhesion mol- ecule gene transcription and subsequent PMN adhesion. Vet Immunol Immunopathol 112: 272–283. Taubert, A., Zahner, H., and Hermosilla, C. (2006b) Dynam- ics of transcription of immunomodulatory genes in endot- helial cells infected with different coccidian parasites. Vet Parasitol 142: 214–222. Toborek, M., Lee, Y.W., Flora, G., Pu, H., Andras, I.E., Wyle- gala, E., et al. (2005) Mechanisms of the blood–brain barrier disruption in HIV-1 infection. Cell Mol Neurobiol 25: 181–199. Tripathi, A.K., Sullivan, D.J., and Stins, M.F. (2006) Plasmo- dium falciparum-infected erythrocytes increase intercellu- lar adhesion molecule 1 expression on brain endothelium through NF-kappaB. Infect Immun 74: 3262–3270. Tripathi, A.K., Sullivan, D.J., and Stins, M.F. (2007) Plasmo- dium falciparum-infected erythrocytes decrease the integ- rity of human blood–brain barrier endothelial cell monolayers. J Infect Dis 195: 942–950. Tripathi, A.K., Sha, W., Shulaev, V., Stins, M.F., and Sullivan, D.J., Jr (2009) Plasmodium falciparum-infected erythro- cytes induce NF-kappaB regulated inflammatory pathways in human cerebral endothelium. Blood 114: 4243–4252. Turner, G.D., Morrison, H., Jones, M., Davis, T.M., Looaree- suwan, S., Buley, I.D., et al. (1994) An immunohistochemi- cal study of the pathology of fatal malaria. Evidence for widespread endothelial activation and a potential role for intercellular adhesion molecule-1 in cerebral sequestra- tion. Am J Pathol 145: 1057–1069. Verma, S., Nakaoke, R., Dohgu, S., and Banks, W.A. (2006) Release of cytokines by brain endothelial cells: a polarized response to lipopolysaccharide. Brain Behav Immun 20: 449–455. Wang, X., Michie, S.A., Xu, B., and Suzuki, Y. (2007) Impor- tance of IFN-gamma-mediated expression of endothelial VCAM-1 on recruitment of CD8+ T cells into the brain during chronic infection with Toxoplasma gondii. J Interferon Cytokine Res 27: 329–338. Wassmer, S.C., Moxon, C.A., Taylor, T., Grau, G.E., Molyneux, M.E., and Craig, A.G. (2011) Vascular endothe- lial cells cultured from patients with cerebral or uncompli- cated malaria exhibit differential reactivity to TNF. Cell Microbiol 13: 198–209. Weiss, J.M., Nath, A., Major, E.O., and Berman, J.W. (1999) HIV-1 Tat induces monocyte chemoattractant protein-1- mediated monocyte transmigration across a model of the human blood–brain barrier and up-regulates CCR5 expres- sion on human monocytes. J Immunol 163: 2953–2959. Weksler, B.B., Subileau, E.A., Perriere, N., Charneau, P., Holloway, K., Leveque, M., et al. (2005) Blood–brain barrier-specific properties of a human adult brain endothe- lial cell line. FASEB J 19: 1872–1874. Workman, J.L., Weber, M.D., and Nelson, R.J. (2011) Dietary arginine depletion reduces depressive-like responses in male, but not female, mice. Behav Brain Res 223: 81– 87. Xia, M., and Hyman, B.T. (2002) GROalpha/KC, a chemokine receptor CXCR2 ligand, can be a potent trigger for neu- ronal ERK1/2 and PI-3 kinase pathways and for tau hyperphosphorylation-a role in Alzheimer’s disease? J Neuroimmunol 122: 55–64. Zhang, Y., and Barres, B.A. (2010) Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr Opin Neu- robiol 20: 588–594. Zhang, R., Miller, R.G., Gascon, R., Champion, S., Katz, J., Lancero, M., et al. (2009) Circulating endotoxin and sys- temic immune activation in sporadic amyotrophic lateral sclerosis (sALS). J Neuroimmunol 206: 121–124. Zlokovic, B.V. (2008) The blood–brain barrier in health and chronic neurodegenerative disorders. Neuron 57: 178– 201. 1478 D. J. Grab, S. J. Chakravorty, H. van der Heyde and M. F. Stins © 2011 Blackwell Publishing Ltd, Cellular Microbiology, 13, 1470–1478