ArticlePDF Available

The Chemokine Receptor CCR2 Mediates the Binding and Internalization of Monocyte Chemoattractant Protein-1 along Brain Microvessels

Authors:

Abstract and Figures

Previous results from this laboratory revealed the presence of high-affinity saturable binding sites for monocyte chemoattractant protein-1 (MCP-1) along human brain microvessels (Andjelkovic et al., 1999; Andjelkovic and Pachter, 2000), which suggested that CC chemokine receptor 2 (CCR2), the recognized receptor for this chemokine, was expressed by the brain microvascular endothelium. To test the role of CCR2 directly in mediating MCP-1 interactions with the brain microvasculature, we assessed MCP-1 binding activity in murine brain microvessels isolated from wild-type mice and from CCR2 (-/-) mice engineered to lack this receptor. Results demonstrate that MCP-1 binding is greatly attenuated in microvessels prepared from CCR2 (-/-) mice compared with wild-type controls. Moreover, microvessels from wild-type mice exhibited MCP-1-induced downmodulation in MCP-1 binding and a recovery of binding activity that was not dependent on de novo protein synthesis. Furthermore, MCP-1 was shown to be internalized within wild-type microvessels, but not within microvessels obtained from CCR2 (-/-) mice, additionally demonstrating that CCR2 is obligatory for MCP-1 endocytosis. Last, internalization of MCP-1, but not transferrin, was observed to be inhibited by disruption of caveolae. Internalized MCP-1 also colocalized at some sites with caveolin-1, a major protein of caveolae, implying that this chemokine is endocytosed, in part, via nonclathrin-coated vesicles. These results prompt consideration that MCP-1 signals may be relayed across the blood-brain barrier by highly specialized interactions of this chemokine with its cognate receptor, CCR2, along brain microvascular endothelial cells.
Internalization of MCP-1 binding sites along murine brain mi- crovessels. Top , Microvessels were re- acted with biot.-rmMCP-1 at 37 ° C, fi xed with 4% paraformaldehyde, and then ei- ther reacted directly with avidin- fl uorescein (nonpermeabilized) or per- meabilized with Tween 20 before reaction with avidin- fl uorescein, as de- scribed in Materials and Methods. Com- pared with Figure 1, wild-type microves- sels ( WT ) that were reacted with labeled chemokine at 37 ° C and that were not permeabilized reveal a greatly attenuated signal. Permeabilization, however, re- stores the detection of biot.-rmMCP-1, suggesting that the labeled chemokine had been internalized within wild-type microvessels at the elevated temperature. Microvessels from CCR2 ( Ϫ / Ϫ ) mice, in contrast, demonstrated only a weak signal regardless of whether or not they had been permeabilized, also implying that they do not internalize MCP-1. Scale bar, 50 ␮ m. Bottom , Three-dimensional ren- derings of microvessels from wild-type mice depicting the distribution of biot.- rmMCP-1 relative to the endothelial plasma membrane. In accordance with the procedures that were described in Materials and Methods, the microvessels were reacted with biot.-rmMCP-1 at ei- ther 4 or 37 ° C and then were fi xed and permeabilized. Next the samples were stained consecutively with avidin- fl uorescein to reveal chemokine localiza- tion ( green ) and then with rho.-WGA to indicate endothelial plasma membrane ( red ). Samples were subject to confocal microscopy and three-dimensional rendering as described in Materials and Methods, and the images are oriented so that viewer is looking “ on face ” toward the microvascular lumen. Chemokine staining clearly lies external to the plasma membrane at 4 ° C and internal to it at 37 ° C (some chemokine staining actually might re fl ect complete transit into the lumen at the elevated temperature).
… 
Content may be subject to copyright.
The Chemokine Receptor CCR2 Mediates the Binding and
Internalization of Monocyte Chemoattractant Protein-1 along
Brain Microvessels
Kirk A. Dzenko,
1
Anuska V. Andjelkovic,
1
William A. Kuziel,
2
and Joel S. Pachter
1
1
Blood–Brain Barrier Laboratory, Department of Pharmacology, University of Connecticut Health Center, Farmington,
Connecticut 06030, and
2
Department of Molecular Genetics and Microbiology and Institute for Cellular and Molecular
Biology, University of Texas, Austin, Texas 78712
Previous results from this laboratory revealed the presence of
high-affinity saturable binding sites for monocyte chemoattrac-
tant protein-1 (MCP-1) along human brain microvessels (And-
jelkovic et al., 1999; Andjelkovic and Pachter, 2000), which
suggested that CC chemokine receptor 2 (CCR2), the recog-
nized receptor for this chemokine, was expressed by the brain
microvascular endothelium. To test the role of CCR2 directly in
mediating MCP-1 interactions with the brain microvasculature,
we assessed MCP-1 binding activity in murine brain microves-
sels isolated from wild-type mice and from CCR2 (/) mice
engineered to lack this receptor. Results demonstrate that
MCP-1 binding is greatly attenuated in microvessels prepared
from CCR2 (/) mice compared with wild-type controls.
Moreover, microvessels from wild-type mice exhibited MCP-1-
induced downmodulation in MCP-1 binding and a recovery of
binding activity that was not dependent on de novo protein
synthesis. Furthermore, MCP-1 was shown to be internalized
within wild-type microvessels, but not within microvessels ob-
tained from CCR2 (/) mice, additionally demonstrating that
CCR2 is obligatory for MCP-1 endocytosis. Last, internalization
of MCP-1, but not transferrin, was observed to be inhibited by
disruption of caveolae. Internalized MCP-1 also colocalized at
some sites with caveolin-1, a major protein of caveolae, imply-
ing that this chemokine is endocytosed, in part, via nonclathrin-
coated vesicles. These results prompt consideration that
MCP-1 signals may be relayed across the blood–brain barrier
by highly specialized interactions of this chemokine with its
cognate receptor, CCR2, along brain microvascular endo-
thelial cells.
Key words: MCP-1; receptors; brain; microvessels; blood–
brain barrier; endothelial cells
Chemokines, which are specialized cytokines, belong to a super-
family of proteins that is composed of four recognized classes
known as
,
,
, and
, or CXC, CC, C, and CXXXC, respec-
tively, by which C refers to conserved cysteine residues, and X
refers to the number of amino acids between cysteines (Rossi and
Zlotnick, 2000). As their name implies, chemokines act as che-
motactic factors that direct the emigration of leukocytes to select
tissue locales during inflammatory episodes. The
chemokine
monocyte chemoattractant protein-1 (MCP-1), newly termed
CCL2 (Rossi and Zlotnick, 2000), has been implicated in medi-
ating the infiltration of mononuclear leukocytes into the CNS
during a variety of neuroinflammatory conditions (Ghirnikar et
al., 1998; Miller and Meucci, 1999; Huang et al., 2000). However,
the mechanism or mechanisms by which this chemokine effects
extravasation into the CNS remain unclear. Confounding this
issue is that astrocytes, which project their foot processes onto the
microvessels comprising the blood–brain barrier (BBB; Lass-
mann et al., 1991), are the predominant source of MCP-1 in
several neuroinflammatory conditions (Ransohoff et al., 1993;
Berman et al., 1996; Glabinski et al., 1996; Van Der Voorn et al.,
1999; Sauder et al., 2000; Sharafeldin et al., 2000). That MCP-1
primarily originates from a source located behind the BBB begs
the following question: How does MCP-1 reach leukocytes resid-
ing in the microvessel lumen? Because chemokines might guide
cells other than leukocytes to the neural parenchyma (Rezaie and
Male, 1999; Silverman et al., 2000), the significance of this query
extends beyond matters related solely to inflammation.
Although MCP-1 might reach intravascular leukocytes by dif-
fusing between endothelial cells, this avenue is likely to be
restricted severely by high-resistance tight junctions of the BBB
(Rubin and Staddon, 1999; Kniesel and Wolburg, 2000). In fact,
other
chemokines, e.g., macrophage inflammatory protein-1
(MIP-1
) and MIP-1
, undergo only minimal passage via this
route (Banks and Kastin, 1996). Alternatively, MCP-1 might bind
to the abluminal endothelial surface and then be conveyed by
transcytosis to the luminal side and/or stimulate a signal trans-
duction event that fosters leukocyte extravasation. Consistent
with these possibilities, the
chemokine interleukin-8 (IL-8) has
been reported to undergo abluminal-to-luminal to transcytosis
across dermal microvessels (Middleton et al., 1997), and BBB
endothelial cells harbor highly selective transport systems for the
vectorial movement of cytokines and other ligands into and out of
the brain (Banks and Kastin, 1991; Friden, 1993; Gutierrez et al.,
1993, 1994; Banks et al., 1994; Abbott and Romero, 1996; De-
houck et al., 1997; Makic et al., 1998; Rose and Audus, 1998).
Moreover, cytokines evoke adhesion molecules (Dobbie et al.,
1999; Wong et al., 1999) and heighten permeability (Megyeri et
Received March 19, 2001; revised July 20, 2001; accepted Sept. 10, 2001.
This work was supported in part by National Institute of Mental Health Grant
1RO1-MH54718-01A1 and National Multiple Sclerosis Society Grant RG2633-A-
1/3 to J.S.P.
Correspondence should be addressed to J. S. Pachter, Blood–Brain Barrier Labo-
ratory, Department of Pharmacology, University of Connecticut Health Center, 263
Farmington Avenue, Farmington, CT 06030. E-mail: pachter@nso1.uchc.edu.
Copyright © 2001 Society for Neuroscience 0270-6474/01/219214-10$15.00/0
The Journal of Neuroscience, December 1, 2001, 21(23):9214–9223
al., 1992; Deli et al., 1995) at the BBB, conditions that could
predispose toward leukocyte extravasation.
Functional binding of MCP-1 along the BBB likely would
require the expression of selective receptors for this chemokine.
Recently, this laboratory identied separate, high-afnity binding
sites for MCP-1 along the abluminal surface of endothelial cells
within human brain microvessels (Andjelkovic et al., 1999; And-
jelkovic and Pachter, 2000). Although these sites resemble CCR2,
the G-protein-coupled receptor (GPCR) for MCP-1 that is
present on leukocytes and other hemopoietic cells (Rossi and
Zlotnick, 2000), direct evidence of CCR2 participation in MCP-1
interactions with brain microvessels has not been conrmed.
Because other putative receptors for MCP-1 have been proposed
(Boddeke et al., 2000), there is a need to clarify the role of CCR2
in MCP-1 binding at the BBB. Consequently, efforts were under-
taken here to test the hypothesis that CCR2 is obligatory for
MCP-1 binding and postbinding events along brain microvessels.
Results indicate that such vessels from mice lacking CCR2 do not
exhibit these activities. In addition, internalization of MCP-1
apparently occurs partly via a caveolae-dependent process. Be-
cause caveolae participate in both signal transduction and trans-
port (Shaul and Anderson, 1998; Feng et al., 1999), engagement
of MCP-1 by CCR2 on the abluminal microvascular surface may be
critical in relaying the signal of this chemokine across the BBB.
MATERIALS AND METHODS
Mice. Mice lacking CCR2, i.e., CCR2 (/), were generated by homol-
ogous recombination as reported previously (Kuziel et al., 1997). Both
CCR2 (/) and wild-type mice were of the genetic background
C57BL/6J129P3/J.
Isolation of murine microvessels. Animals were killed with CO
2
in
accordance with measures stipulated by the Animal Care and Use Guide-
lines of the University of Connecticut Health C enter (Animal Welfare
Assurance A3471-01). Immediately after death, craniotomy was per-
formed, and the entire cerebrum was removed and dissected free of
meninges. Microvessels were isolated from brain tissue by the procedure
detailed in recent publications (Andjelkovic et al., 1999; Andjelkovic and
Pachter, 2000). Briey, this procedure involved initially separating mi-
crovessels from parenchymal tissue by gentle Dounce homogenization
and then pressing the tissue extract through graded sieves of smaller
porosity to remove macrovascular segments. Microvessels subsequently
were isolated from the dissociated material by centrifugation through
sequential dextran and Percoll gradients. Because enzymatic treatment
was avoided during the isolation protocol, the basement membrane
surrounding the resultant microvascular fragments was left relatively
intact. Isolated microvessels were washed in PBS, pH 7.4, and were used
immediately.
Binding e xperiments. Chemokine binding to brain microvessels was
conducted as described previously (Andjelkovic et al., 1999; Andjelkovic
and Pachter, 2000). In brief, puried microvessels were reacted with
biotinylated recombinant murine (biot.-rm) MCP-1 or biot.-rmMI P-1
(Fluorokine kits, R&D Systems, Minneapolis, MN) in the absence or
presence of increasing concentrations of unlabeled rmMCP-1 (R&D
Systems) or unlabeled rmMIP-1
(Peprotech, Norwood, MA) at speci-
ed temperatures. In some experiments the microvessels were reacted
with biotinylated transferrin or biotinylated cholera toxin subunit B (both
from Sigma, St. Louis, MO). After 2 hr of incubation with biotinylated
ligands the microvessels were reacted with avidin-uorescein (R&D
Systems) for an additional 1 hr at 4°C. For negative controls the mi-
crovessels were incubated with the irrelevant biotinylated soybean tryp-
sin inhibitor (R&D Systems). Samples were viewed with a Zeiss LSM
410 confocal microscope (Oberkochen, Germany) equipped with an
argonkrypton laser [excitation at 488 nm; emission at 515 nm (long-pass
lter)], and images were obtained and processed with Adobe Photoshop
3.0 software (Adobe Systems, San Jose, CA) as detailed previously
(Andjelkovic et al., 1999; Andjelkovic and Pachter, 2000). To quantify
the extent of labeled chemokine binding, we recorded the values of mean
pixel intensity from a total of 25 randomly chosen areas (192 pixels each)
along at least 10 individual microvessels from each sample. Mean pixel
values were obtained similarly from negative controls, and an average
value of this parameter was subtracted from each of the 25 pixel inten-
sities obtained from all of the different chemokine binding conditions.
This correction served to remove background noise caused by indis-
criminate avidin-uorescein binding. Corrected pixel intensities then
were averaged, with the resulting value representing the relative degree
of specic chemokine binding along the microvessels.
To detect internalized ligands, we xed the microvessels in 4% para-
formaldehyde immediately after incubation with biotinylated chemokine,
transferrin, or cholera toxin and then permeabilized them by incubation
in PBS containing 0.5% (w/v) Tween 20 (Sigma). Then the ligands were
revealed by subsequent reaction with avidin-uorescein.
Ligand-induced downmodulation/recovery of MCP-1 binding sites. To
assess the loss of biot.-rmMCP-1 surface binding because of downmodu-
lation, we preexposed the microvessels to unlabeled rmMCP-1 (2.5
g/ml) for varying times at 37°C. Next the samples were washed with
PBS and subjected to the standard binding assay by using biot.-rmMCP-
1/avidin-uorescein at 4°C.
For evaluation of the recovery of biot.-rmMCP-1 binding after down-
modulation, the microvessels were incubated rst with unlabeled
rmMCP-1 (2.5
g/ml) for 2 hr at 37°C. Then the samples were washed
with PBS and allowed to recover for varying periods of time in PBS
containing 10 m
MD-glucose (Sigma) at 37°C, after which the standard
binding assay was performed. To determine whether the reexpression of
MCP-1 binding activity along the brain microvessel surface required de
novo protein synthesis, we exposed the microvessels to 10
g/ml cyclo-
heximide (Sigma) throughout the recovery phase. Specically, cyclohex-
imide was introduced during the last 30 min of exposure of the microves-
sels to unlabeled rmMCP-1. At the end of this period the microvessels
were washed with PBS containing cycloheximide and then resuspended
in fresh PBS/cycloheximide/
D-glucose and incubated at 37°C for 90 min
(recovery phase). The standard binding assay was conducted after this
recovery phase.
Filipin III treatment. To assess the contribution of caveolae to the
process of MCP-1 internalization, we added lipin III (5
g/ml; Sigma)
to the microvessels for a 30 min pretreatment before the addition of
biotinylated ligand. Then the microvessels were washed with PBS, and
binding reactions were performed in the absence of lipin III. Exposure
to lipin III was minimized intentionally to avoid toxicity, and the
microvessels remained viable throughout this protocol as judged by
trypan blue exclusion.
Heparinase treatment. So that the role of heparan sulfate in MCP-1
binding could be investigated, the microvessels were digested with 176
U/ml Heparinase I (Sigma) for 45 min at 37°C. This procedure served to
remove all immunodetectable heparan sulfate but did not affect adversely
the gross integrity of the microvessels (Andjelkovic et al., 1999). After
digestion the samples were washed with PBS and were submitted to the
standard binding assay.
Histochemistry/immunoc ytochemistry. To gauge the distribution of
bound/internalized biot.-rmMCP-1 relative to the endothelial plasma
membrane, we xed the microvessels in 4% paraformaldehyde, perme-
abilized them after reaction with chemokine, and then costained them
with rhodamine-conjugated lectin wheat germ agglutinin (rho.-WGA;
Vector Laboratories, Burlingame, CA) at 5
g/ml.
To assess the colocalization of internalized MCP-1 with caveolin-1, we
reacted the microvessels with biot.-rmMCP-1 at 37°C for 10 min, which
was sufcient time to observe chemokine transport across the endothelial
plasma membrane (as judged separately by rho.-WGA staining). This
time point thus was considered appropriate to maximize the detection of
internalized MCP-1 with caveolin-1-containing vesicles at the earliest
stages of their invagination. After incubation with chemokine the mi-
crovessels were xed with 4% paraformaldehyde and permeabilized.
Then the samples were reacted with Alexa Fluor 488-conjugated avidin
(Molecular Probes, Eugene, OR) at 5
g/ml. The reason for using the
Alexa Fluor 488-derivative here, instead of uorescein-conjugated avi-
din, is that the former uorophore has a higher quantum yield. Because
the signal associated with transported biot.-rmMCP-1 at this early time
point was low, this necessitated the use of the brighter uorophore. After
reaction with Alexa 488 the samples were rinsed in PBS and incubated
with polyclonal anti-caveolin-1 antibody (2
g/ml; Transduction Labs,
Lexington, KY). Next the microvessels were washed again in PBS and
reacted with Texas Red-conjugated goat anti-rabbit IgG (10
g/ml;
Vector Laboratories). Lectin and anti-caveolin-1-stained samples were
viewed with a Zeiss 410 LS confocal microscope, using the after-lter
congurations uorescein and Alexa Fluor 488 [excitation at 488 nm,
emission at 515540 nm (bandpass lter)], rhodamine [excitation at 568
Dzenko et al. CCR2 on Brain Microvessels J. Neurosci., December 1, 2001, 21(23):92149223 9215
nm, emission at 590 nm (long-pass lter)], and Texas Red [excitation at
568 nm, emission at 610 nm (long-pass lter)].
Three-dimensional renderings of biot.rmMCP-/rho.-WGA costained
microvessels were generated from a z-series of confocal images through
the entirety of microvessel samples, using VoxelView software (Vital
Images, Faireld, IA) as described previously (Andjelkovic et al., 1999;
Andjelkovic and Pachter, 2000).
Statistical analysis. To determine the signicance of the effects of
varied treatments on biot.rm-MCP-1 binding to brain microvessels, we
performed a one-way ANOVA, followed by a Bonferroni multiple com-
parisons test.
RESULTS
MCP-1 binding along brain microvessels
Figure 1 shows the binding of biot.-rmMCP-1 along the abluminal
surface of isolated murine brain microvessels. In microvessel
samples prepared from wild-type mice, the distribution of labeled
MCP-1 appears to be relatively continuous along the microvas-
cular surface, as has been described recently for the correspond-
ing human chemokine derivative along human brain microvessels
(Andjelkovic et al., 1999). Only negligible signal was detected in
samples exposed to biot.-soybean trypsin inhibitor (negative con-
trol), reecting the specicity of the binding reaction. Heparan
sulfate moieties along either the endothelial surface or attendant
basement membrane were not major factors in biot.-rmMCP-1
binding, because digestion of the microvessels with heparinase I
[sufcient to eliminate all heparan sulfate immunocytochemical
reactivity (Andjelkovic et al., 1999; data not shown)] failed to
eliminate the chemokine signal and only very slightly depressed it.
Thus, although heparan sulfate can bind MCP-1, albeit with an
afnity 23 log units less than CCR2 (Hoogewerf et al., 1997), it
is not the primary binding domain for this chemokine on brain
microvessels under the conditions that were used. Because these
new ndings recapitulated those previously observed with human
microvessels, it established the murine system as a suitable para-
digm to investigate the role of CCR2 in mediating MCP-1 inter-
actions at the human BBB.
In marked contrast to the situation observed with wild-type
vessels, signicantly diminished binding of biot.-rmMCP-1 was
detected on brain microvessels from CCR2 (/) mice. What-
ever little binding was observed in the CCR2 (/) samples may
derive, at least partially, from heparan sulfate, because hepari-
nase digestion nearly completely eradicated the detectable signal.
Importantly, binding of biot.-rmMIP-1
was nearly invariant
between microvessels from wild-type and CCR2 (/) mice,
exhibiting a similar punctate pattern previously observed with
human tissue (Andjelkovic et al., 1999; Andjelkovic and Pachter,
2000) and indicating that CCR2 ablation generally did not affect
chemokine binding. These ndings provide the rst direct evi-
dence that the binding of MCP-1 along brain microvessels is
linked inextricably to the expression of CCR2.
Additional evidence supporting the assertion that CCR2 medi-
ates MCP-1 binding to brain microvessels was provided by a
competitive ligand-binding assay (Fig. 2). As described recently
for human brain microvessels (Andjelkovic et al., 1999; Andjel-
kovic and Pachter, 2000), the binding of biot.-rmMCP-1 could be
antagonized by its unlabeled homolog, but not by unlabeled
rmMIP-1
, the latter a ligand for chemokine receptors CCR1
and CCR5, but not for CCR2. The residual binding exhibited in
the presence of competitor ligand, as for the case with brain
microvessels from CCR2-decient mice, might reect the small
contribution from nonreceptor-mediated association of biotinyl-
ated chemokine with heparan sulfate and/or other glycosamino-
glycans (GAGs; Hoogewerf et al., 1997; Kuschert et al., 1999).
Figure 1. Biotinylated chemokine
binding along murine brain microves-
sels. Top, Representative examples of
uorescent detection of chemokines at-
tached to the brain microvascular sur-
face. Detection of biot.-rmMCP-1 and
biot.-rmMI P-1
binding was performed
at 4°C as described in Materials and
Methods. Diminished binding of biot.-
rmMCP-1 to microvessels from CCR2
(/) mice, compared with those from
wild-type mice (WT), is readily appar-
ent. No such difference between these
respective microvessel populations is
observed for biot.-rmMIP-1
binding,
and heparinase treatment failed to ab-
rogate biot.-rmMCP-1 binding. Scale
bar, 50
m. Bottom, Quantitative analy-
sis of biotinylated chemokine binding.
Mean pixel intensities SEM, reect-
ing relative chemokine binding, were
determined as described in Materials
and Methods and were corrected for
background noise by subtraction of in-
tensity values associated with negative
controls. Values represent those deter-
mined from at least three different ex-
periments. *p 0.001 when contrasted
with corresponding wild-type value.
9216 J. Neurosci., December 1, 2001, 21(23):92149223 Dzenko et al. CCR2 on Brain Microvessels
Downmodulation and recovery of MCP-1 binding
Because CCR2 on leukocytes and transfected cells has been
reported to undergo downmodulation in response MCP-1 expo-
sure (Sarau et al., 1997; Aragay et al., 1998; Fantuzzi et al., 1999),
it was reasoned that if the interaction of this chemokine with
brain microvessels were to be mediated predominantly or exclu-
sively by CCR2, then preexposure to unlabeled MCP-1 should
lessen the degree of biot.-rmMCP-1 binding along the microvas-
cular endothelial surface. Figure 3 shows that, when microvessels
were preexposed to unlabeled MCP-1 for varying lengths of time,
washed free of unbound chemokine, and then reexposed to biot.-
rmMCP-1, the degree of labeled MCP-1 binding to the microves-
sel surface gradually decreased. A nearly 50% attenuation in
maximal binding (i.e., the signal detected when there was no
preexposure to unlabeled ligand) was achieved after 15 min of
preexposure to unlabeled MCP-1. This loss of MCP-1 binding
activity along the abluminal brain microvascular surface could be
recovered within minutes, similar to the time scale reported for
the ligand-induced loss of expression of several chemokine recep-
tors on other cell types (Madani et al., 1998; Feniger-Barish et al.,
1999). In this case the recovery of 50% of maximal biot.-
rmMCP-1 binding was attained within 30 min, and complete
recovery was attained within 60 min after removal of the unla-
beled chemokine. It was determined further that the restoration
of MCP-1 binding was not dependent on de novo protein synthe-
sis, because recovery proceeded unabated in the presence of
cycloheximide (Fig. 4). These features are consistent with the
interpretation that, like other chemokine receptors stimulated by
their respective ligands, MCP-1 binding sites are internalized and
then recycled to the endothelial cell surface as a consequence of
MCP-1 exposure (Mack and Schlondorff, 2000).
Internalization of MCP-1
To conrm whether MCP-1 is, in fact, internalized after binding
to the brain microvascular surface, we performed the binding of
biot.-rmMCP-1 at 37°C, followed by interaction with avidin-
uorescein at 4°C. Figure 5, top, indicates that under these con-
ditions the detection of biot.-rmMCP-1 on the microvascular
surface was reduced signicantly compared with that seen when
chemokine binding was performed at 4°C. However, when mi-
crovessels incubated with biot.-rmMCP-1 at 37°C were xed and
then permeabilized before interaction with avidin-uorescein,
the uorescent signal was detected within the endothelial cell
cytoplasm. The staining pattern revealed by permeabilization
also appeared to be more patchy than that observed with
samples exposed to chemokine at 4°C (also compare with Fig. 1).
These results are taken to indicate that biotinylated chemokine
Figure 2. Competition of biotinylated MCP-1 binding along murine
brain microvessels. Competition studies were performed at 4°C with a
constant concentration of biot.-rmMCP-1 and increasing concentrations
of unlabeled chemokines (indicated by symbols). Binding was quantitated
as described in Materials and Methods and is reported as the percent-
age SEM of maximal binding achieved in the absence of inhibitor
ligand. Values represent those determined from at least three different
experiments.
Figure 3. Loss and recovery of MCP-1 binding sites on murine brain
microvessels after ligand exposure. Microvessels were reacted with unla-
beled MCP-1 at 37°C for 2 hr (recovery) or for varying periods of time
(loss). After exposure to unlabeled chemokine the microvessels were
washed in PBS and either were exposed immediately to biot.-rmMC P-1
for2hrat4°C (loss) or were incubated at 37°C for varying periods of time
and then exposed to biot.-rmMCP-1 (recovery). After reaction with
biot.-rmMCP-1 the samples were subjected to the standard binding assay
conditions, and binding intensity along the microvessels was analyzed as
described in Materials and Methods. The extent of both loss and recovery
of biot.-rmMCP-1 binding is indicated as the percentage SEM of
maximal binding obtained in the absence of any previous exposure of the
microvessels to unlabeled MCP-1. Values represent those determined
from at least three different experiments.
Figure 4. Effect of protein synthesis inhibition on the recovery of MCP-1
binding sites along murine brain microvessels. Microvessels were exposed
to unlabeled MCP-1 as described in Figure 3, except that in the last 30
min the protein synthesis inhibitor cycloheximide (10
g/ml) either was
added to the incubation mixture or was not. After chemokine exposure
the microvessels treated with cycloheximide were allowed to recover for
90 min in the continued presence of protein synthesis inhibitor while the
control samples continued their recovery in the absence of cycloheximide.
Binding was quantitated as described in Materials and Methods and is
reported as the percentage SEM of maximal binding obtained in the
absence of any previous exposure of the microvessels to unlabeled MCP-1.
Values represent those determined from three different experiments.
Dzenko et al. CCR2 on Brain Microvessels J. Neurosci., December 1, 2001, 21(23):92149223 9217
had been internalized at the elevated temperature and was not
available to interact with the avidin-uorophore in the nonper-
meabilized condition. This interpretation is supported in Figure
5, bottom, which depicts three-dimensional renderings of mi-
crovessels and the relative distribution of labeled MCP-1 to the
endothelial surface. In this case the combined visualization of
biot.-rmMCP-1 and rho.-WGA, the latter marking endothelial
plasma membrane (Vorbrodt et al., 1994), reveals the chemokine
to lie external to the membrane at 4°C and internal to it at 37°C.
To establish that internalization of biot.-rmMCP-1 most likely
is mediated by CCR2, we also performed an identical binding
paradigm at 37°C with brain microvessels from CCR2 (/)
mice. In this case there was little signal detected in the cytoplasm
of either permeabilized or nonpermeabilized vessels. In fact,
biot.-rmMCP-1 staining of these two populations of vessels did
not differ much from each other or from microvessels of
CCR2-decient mice exposed to MCP-1 at 4°C, also demon-
strating that non-CCR2 binding sites for MCP-1 are not sub-
ject to internalization.
Last, the mechanism by which MCP-1 is internalized along
brain microvessels was investigated. Two pathways for GPCR-
mediated internalization of agonist ligands currently are recog-
nized: one using clathrin-coated pits (Bohm et al., 1997; Lefko-
witz, 1998) and the other using nonclathrin-coated plasmalemmal
vesicles called caveolae (Anderson, 1998; Okamoto et al., 1998).
However, several facts regarding caveolae, along with the need to
reconcile how MCP-1 deposited in the perivascular space can
exert its effect on leukocytes on the other side of the BBB, lead us
to focus on the caveolar pathway at this time: (1) caveolae are
particularly prominent in endothelial cells, including those of
brain microvessels (Ikezu et al., 1998); (2) caveolae mediate the
vectorial movement of low-density lipoprotein across cultured
endothelial cells derived from brain microvessels (Dehouck et al.,
1997); (3) caveolae have been suggested to be involved possibly in
the abluminal-to-luminal transcytosis of the
chemokine IL-8
across dermal venules (Middleton et al., 1997); and (4) caveolae
have been implicated in signal transduction events (Lisanti et al.,
1994; Shaul and Anderson, 1998). Thus several precedents al-
ready have been set highlighting the possibility that caveolae
could relay MCP-1 signals across the BBB. To investigate use of
caveolae in the internalization process, we pretreated brain mi-
crovessels with the agent lipin III before their exposure to
labeled MCP-1 at 37°C. Filipin III is a sterol-binding agent that
removes cholesterol from membranes and, thus, selectively dis-
rupts caveolar microdomains (Schnitzer et al., 1994). Figure 6
reveals that previous treatment of microvessels with lipin III
enabled the detection of biot.-rmMCP-1 when samples were
exposed to chemokine at 37°C and then xed, without the need of
permeabilization before the application of avidin-uorescein. In
marked contrast to the near-complete loss of detectable staining
of nonpermeabilized control microvessels, the staining of perme-
abilized and nonpermeabilized lipin III-treated samples ap-
Figure 5. Internalization of MCP-1
binding sites along murine brain mi-
crovessels. Top, Microvessels were re-
acted with biot.-rmMCP-1 at 37°C, xed
with 4% paraformaldehyde, and then ei-
ther reacted directly with avidin-
uorescein (nonpermeabilized) or per-
meabilized with Tween 20 before
reaction with avidin-uorescein, as de-
scribed in Materials and Methods. Com-
pared with Figure 1, wild-type microves-
sels (WT) that were reacted with labeled
chemokine at 37°C and that were not
permeabilized reveal a greatly attenuated
signal. Permeabilization, however, re-
stores the detection of biot.-rmMCP-1,
suggesting that the labeled chemokine
had been internalized within wild-type
microvessels at the elevated temperature.
Microvessels from CCR2 (/) mice, in
contrast, demonstrated only a weak signal
regardless of whether or not they had
been permeabilized, also implying that
they do not internalize MCP-1. Scale bar,
50
m. Bottom, Three-dimensional ren-
derings of microvessels from wild-type
mice depicting the distribution of biot.-
rmMCP-1 relative to the endothelial
plasma membrane. In accordance with
the procedures that were described in
Materials and Methods, the microvessels
were reacted with biot.-rmMCP-1 at ei-
ther4or37°C and then were xed and
permeabilized. Next the samples were
stained consecutively with avidin-
uorescein to reveal chemokine localiza-
tion ( green) and then with rho.-WGA to
indicate endothelial plasma membrane
(red). Samples were subject to confocal
microscopy and three-dimensional rendering as described in Materials and Methods, and the images are oriented so that viewer is looking on face
toward the microvascular lumen. Chemokine staining clearly lies external to the plasma membrane at 4°C and internal to it at 37°C (some chemokine
staining actually might reect complete transit into the lumen at the elevated temperature).
9218 J. Neurosci., December 1, 2001, 21(23):92149223 Dzenko et al. CCR2 on Brain Microvessels
peared to be similar to each other in pattern and intensity.
Particularly noteworthy is that the staining of both lipin III-
treated samples was delineated along the microvascular surface,
distinct from the patchy cytoplasmic pattern manifested by con-
trol samples (Figs. 5, 6). The interpretation here is that, by
disrupting caveolae-based internalization, lipin III treatment
allowed surface-bound biot.-rmMCP-1 to remain on the cell
surface and, thus, accessible to reaction with avidin-uorescein in
the absence of membrane permeabilization.
To preclude the possibility that treatment with lipin III merely
resulted in cellular toxicity or adversely affected all endocytic
processes, we also evaluated the effect of this agent on transferrin
internalization, which is mediated by clathrin-coated vesicles
(Benlimame et al., 1998) (Fig. 7). As was the case with labeled
MCP-1, the detection of biot.-transferrin at 37
C was achieved
only when microvessels were xed and then permeabilized before
exposure to avidin-uorescein. Failure to permeabilize the mi-
crovessels resulted in a lack of biot.-transferrin detection, imply-
ing sequestration of this ligand behind the plasma membrane and
validating that transferrin internalization had occurred. Contrary
to the effects on biot.-rmMCP-1 detection, however, previous
treatment of microvessels with lipin III did not prevent the
diminished detection of the biot.-transferrin signal at 37°C. Both
lipin III-treated and control samples required detergent perme-
abilization to reveal biot.-transferrin bound at 37°C, implying that
exposure to lipin III did not prevent transferrin internalization.
To conrm additionally that the action of lipin III was not
peculiar to MCP-1, we investigated the effect of this agent on the
internalization of cholera toxin, a ligand reported to be endocy-
tosed by endothelial cells via the caveolar route (Orlandi and
Fishman, 1998). In this case (Fig. 7) the lipin III treatment
yielded results identical to those observed with MCP-1: it enabled
the detection of cholera toxin when the samples were exposed to
ligand at 37°C and then xed, without the need of membrane
permeabilization. The interpretation here too is that, by blocking
caveolae-mediated internalization, lipin III caused cholera toxin
to remain on the microvessel surface.
Last, colocalization of internalized MCP-1 with caveolin-1, a
major structural protein of caveolae (Rothberg et al., 1992; Schle-
gel and Lisanti, 2001), was examined by double-label confocal
microscopy to corroborate further an association of internalized
chemokine with the caveolar pathway. Figure 8 shows the pat-
terns of distribution of biot.-rmMCP-1 and caveolin-1 along brain
microvessels after chemokine exposure at 4 and 37°C. At 4°C both
biot.-rmMCP-1 and caveolin-1 staining appeared to be concen-
trated at or near the abluminal microvascular surface, although
segregated from each other. The lack of a signicant cytoplasmic
chemokine signal is in accord with there being little or no inter-
nalization at this temperature, and the distribution of caveolin-1
is consistent with the presence of caveolae at the plasmalemma
(Kurzchalia and Parton, 1999). After exposure at 37°C for 10 min
the distribution of biot.-rmMCP-1 colocalized with that of
caveolin-1 at some sites, as indicated by the yellow uorescent
signal. In addition, cytoplasmic staining of biot.-rmMCP-1 ap-
peared to be more intense at 37°C than at 4°C, compatible with a
Figure 6. Effect of lipin III on internalization of MCP-1 along murine
brain microvessels. To gauge whether MCP-1 internalization may be
mediated by caveolae, we exposed the microvessels to the caveolae-
disrupting agent lipin III before incubation with biot.-rmMCP-1 at 37°C.
Contrary to nontreated controls, the microvessels pretreated with lipin
III did not require permeabilization to enable the detection of labeled
chemokine, suggesting that biot.-rmMCP-1 remained on the cell surface
as a consequence of caveolar disruption. Scale bar, 50
m.
Figure 7. Effect of lipin III on internalization of transferrin and cholera
toxin along murine brain microvessels. Microvessels received () lipin
III treatment, were exposed to biot.-transferrin or biot.-cholera toxin, and
then were processed as described in Figure 6. Both control and lipin
III-treated microvessels required permeabilization to detect biot.-
transferrin, indicating that the process of transferrin internalization pro-
ceeded despite disruption of the caveolae. In contrast, biot.-cholera toxin
could be observed along the surface of lipin III-treated microvessels both
with and without permeabilization, reecting lipin III-mediated inter-
ference with the internalization of this ligand. Scale bar, 50
m.
Dzenko et al. CCR2 on Brain Microvessels J. Neurosci., December 1, 2001, 21(23):92149223 9219
signicantly higher degree of endocytosis occurring at the ele-
vated temperature. Pretreatment of microvessels with lipin III
caused disruption in the pattern of caveolin-1 staining, resulting
in a lessening in intensity along the microvessel contour and a
redistribution to the cytoplasmic compartment. This change is
consistent with that reported by others (John et al., 2001; Rose-
berry and Hosey, 2001) and possibly reects the cytoplasmic
dispersion of caveolin-1 liberated from plasma membrane-
associated caveolae. Collectively, these results suggest that the
caveolar pathway is used in some capacity during the internaliza-
tion of MCP-1 by endothelial cells of brain microvessels.
DISCUSSION
Previous studies from this laboratory have demonstrated the
presence of high-afnity saturable binding sites for MCP-1 along
human brain microvessels and have shown that these sites display
pharmacological and biochemical properties similar to those re-
ported for CCR2 (Andjelkovic et al., 1999; Andjelkovic and
Pachter, 2000). Here we substantiated these ndings with murine
brain microvessels and further established that the expression of
CCR2 by brain microvascular endothelial cells is obligatory for
the manifestation of MCP-1 binding properties. Specically,
biot.-rmMCP-1 bound to the abluminal surface of isolated brain
microvessels from wild-type mice by a process that was inhibited
by unlabeled MCP-1, but not unlabeled MIP-1
, and also was
primarily independent of the presence of heparan sulfate. Such
biot.-rmMCP-1 binding was reduced greatly along microvessels
isolated from CCR2 (/) mice, whereas the binding of biot.-
rmMIP-1
occurred to similar extents under wild-type and
CCR2 knock-out conditions. As also reported for human mi-
crovessels, biot.-rmMCP-1 was internalized within endothelial
cells of isolated murine vessels after binding at 37
C, but endo
-
cytosis of this chemokine was not observed in microvascular
tissue from CCR2 (/) mice. Last, internalization of biot.-
rmMCP-1 and biot.-cholera toxin, but not biot.-transferrin, ap-
parently was prevented by the use of the caveolae-disrupting
agent lipin III. These results collectively argue that CCR2 is
predominantly, if not solely, responsible for MCP-1 binding along
the abluminal brain microvascular surface and that such receptor-
mediated binding leads, in some proportion, to the internaliza-
tion of ligands via a pathway known to be associated with trans-
cytotic and signal transduction events.
That specic biot.-rmMCP-1 binding to murine brain mi-
crovessels was observed is an important validation of similar
results recently reported with human ligands and tissue (Andjel-
kovic et al., 1999; Andjelkovic and Pachter, 2000) and allays the
prospect that this interaction is restricted to human tissue and/or
is attributable to some artifact uniquely associated with the pro-
curement thereof. On the contrary, it points to the expression of
endothelial MCP-1 binding sites as possibly being a common
mammalian feature. Supporting this interpretation are ndings
from other laboratories of the expression of CCR2 as well as
other chemokine receptors by a variety of endothelial cell types
from human, macaque, bovine, and rodent tissues (Edinger et al.,
1997; Rottman et al., 1997; Feil and Augustin, 1998; Gupta et
al., 1998; Sanders et al., 1998; Volin et al., 1998; Andjelkovic
et al., 1999; Berger et al., 1999; Murdoch et al., 1999; Shaw and
Grieg, 1999; Weber et al., 1999; Andjelkovic and Pachter, 2000;
Molino et al., 2000; Salcedo et al., 2000).
Because brain microvessels from CCR2 (/) mice failed to
exhibit the binding of MCP-1 but retained that of MIP-1
, this
report is the rst to link the expression of a specic chemokine
receptor with selective chemokine binding to endothelial cells.
Thus, whereas competitor chemokine binding assays revealed a
pharmacological prole consistent with CCR2 activity along hu-
Figure 8. Colocalization of internalized
MCP-1 with caveolin-1. Microvessels were
pretreated () with lipin III. Then the
samples were exposed to biot.-rmMCP-1
at4or37°C, xed/permeabilized, and
stained to reveal labeled chemokine
( green) and caveolin-1 (red) localization,
as described in Materials and Methods.
Then confocal images were obtained at a
level approximately midway through the
interior of the microvascular samples, re-
vealing the relative distribution patterns
of labeled chemokine and caveolin-1. In
the control sample exposed to biot.-
rmMCP-1 at 37°C, caveolin-1 staining can
be seen concentrated around the periph-
ery of the microvascular segment (arrows),
with chemokine apparently present dif-
fusely in the cytoplasm. Areas of yellow
uorescence (asterisks) indicate presumed
sites of biot.-rmMCP-1/caveolin-1 colocal-
ization. In the lipin III-treated sample
exposed to chemokine at 37°C, no sites of
colocalization are detected readily, and
biot.-rmMCP-1 appears to be conned to
the membrane surface (arrowheads), with
caveolin-1 expression heightened in some
cytoplasmic locales (arrows). Microvessels
exposed to chemokine at 4°C() lipin
III pretreatment also fail to show any areas of biot.-rmMCP-1/caveolin-1 colocalization. These samples also do not demonstrate as strong a cytoplasmic
distribution of labeled chemokine as that observed in the control sample at 37°C but seemingly manifest a more peripheral chemokine staining
(arrowheads), possibly restricted to the membrane surface. As with the samples exposed to chemokine at 37°C, caveolin-1 distribution appears to be
concentrated along the periphery of the microvessel in the control (arrowheads) but is dispersed more cytoplasmically in the lipin III-treated sample.
Arrows, Caveolin-1; arrowheads, biot.-rmMCP-1; asterisks, biot.-rmMCP-1/caveolin-1 colocalization. Scale bar, 10
m.
9220 J. Neurosci., December 1, 2001, 21(23):92149223 Dzenko et al. CCR2 on Brain Microvessels
man brain microvessels (Andjelkovic et al., 1999; Andjelkovic
and Pachter, 2000), results from this report verify the microvas-
cular expression of this receptor and its role in MCP-1 binding.
The reduction in MCP-1 binding associated with CCR2 absence
also lessens the possibility that binding is attributable to another
receptor with CCR2-like properties (Boddeke et al., 2000).
Hence, bearing any unforeseen ablation in the knock-out mice of
a gene encoding such a similarly functioning receptor for MCP-1,
these ndings offer persuasive evidence that CCR2 is singularly
responsible for the binding of MCP-1 along the abluminal surface
of brain microvessels.
In addition to mediating MCP-1 binding, CCR2 is also oblig-
atory for the endocytosis of MCP-1 within endothelial cells, as
was indicated by the inability of brain microvessels from CCR2
(/) mice to internalize this chemokine. Despite some low
degree of MCP-1 binding to heparan sulfate moieties along the
abluminal microvascular surface [in both wild-type and CCR2
(/) mice], ligand engagement at this level does not result in
internalization and probably reects low-afnity interaction of
the chemokine with GAGs. Weaker binding of this latter type
may serve the purpose of concentrating chemokines in the
perivascular space and/or enabling proper chemokine presenta-
tion to CCR2-bearing cells in or entering the brain parenchyma
(Hoogewerf et al., 1997; Kuschert et al., 1999).
The kinetics of MCP-1-induced loss and recovery of murine
microvascular chemokine binding reported here is similar to that
previously described with human brain microvessels (Andjelkovic
et al., 1999; Andjelkovic and Pachter, 2000) and mirrors the time
course for ligand-stimulated downmodulation and reexpression of
chemokine receptors in other systems (Madani et al., 1998;
Feniger-Barish et al., 1999). Given the relatively rapid times that
are required to achieve half-maximal loss or recovery of MCP-1
binding, the lack of effect of protein synthesis inhibition on the
recovery process, and the dependency of internalization on CCR2
expression, ligand-induced alteration in MCP-1 binding along
brain microvessels is likely to occur by receptor-mediated inter-
nalization and recycling.
The ability of lipin III to abrogate internalization of MCP-1
and cholera toxin, but not transferrin, further prompts intriguing
suggestions as to the fate of this chemokine after its binding to
the abluminal surface of brain microvessels. Specically, the col-
lective evidence points to some portion of internalized MCP-1
entering the caveolae. Caveolae are membrane specializations
enriched in G-protein-coupled receptors, heterotrimeric GTP
binding proteins, IP
3
receptor-like protein, Ca
2
ATPase, and
several protein kinase C isoforms (Isshiki and Anderson, 1999)
and are recognized to transport molecules across endothelial cells
(Anderson, 1998). As such, the association of MCP-1 with this
membrane system may intimate that this chemokine stimulates a
signal transduction pathway or pathways and/or is transferred to
the luminal surface in the process of effecting its actions at the
BBB. Both types of responses have, in fact, been demonstrated in
endothelial cells after exposure to IL-8 (Middleton et al., 1997;
Schraufstatter et al., 2001). Interestingly, previous reports regard-
ing ligand-induced internalization of other chemokine receptors
have described only the use of the clathrin-dependent pathway in
nonvascular cell types (Amara et al., 1997; Mack et al., 1998;
Yang et al., 1999). Our ndings thus might suggest a propensity
for endothelial cells, and possibly those of brain microvessels in
particular, to use caveolae conditionally. Indeed, such a charac-
teristic would be consistent with the expression of highly selective
transporter systems at the BBB (Banks and Kastin, 1991; Friden,
1993; Gutierrez et al., 1993, 1994; Banks et al., 1994; Abbott and
Romero, 1996; Dehouck et al., 1997; Makic et al., 1998; Rose and
Audus, 1998). This interpretation is not meant to exclude a role
for the clathrin-dependent pathway in the internalization of che-
mokines and their receptors at the brain microvascular endothe-
lium but only to impart recognition of the caveolar pathway. Both
endocytic venues, in fact, might operate routinely in the action of
MCP-1 along the brain microvascular endothelium, with the
relative involvement of each depending on factors including cel-
lular cholesterol oxidation state (Okamoto et al., 2000) and/or
degree of receptor phosphorylation (Roettger et al., 1995). Dual
pathways of internalization have, in fact, been suggested for
several peptide ligands including growth hormone (Lobie et al.,
1999), insulin (King and Johnson, 1985; Schnitzer et al., 1994),
and cholecystokinin (Roettger et al., 1995).
Although the physiological relevance of MCP-1 binding/inter-
nalization along brain microvessels remains to be ascertained, it
raises the prospect that leukocyte extravasation into the CNS
might involve chemokine engagement along the abluminal micro-
vascular surface. In what may be viewed as an analogous situation
to the perivascular deposition of MCP-1 by astrocytes in vivo, the
application of MCP-1 to the basolateral surface (akin to the
abluminal surface in vivo), but not to the apical surface (repre-
senting the luminal surface in vivo), of cultured brain microvas-
cular endothelial cells (BMEC) has been observed to stimulate
monocyte transendothelial migration (Andjelkovic et al., 2001).
Assuming there is minimal paracellular leakage of MCP-1 be-
tween the BMEC, which retain their highly restrictive BBB
phenotype under the conditions that have been assayed (Biegel et
al., 1995), a seemingly a posteriori conclusion is that the primary
site of MCP-1 action is along the basolateral BMEC surface,
possibly at CCR2. It thus is intriguing to speculate that reduced
sensitivity to experimental autoimmune encephalomyelitis
(EAE) in CCR2 (/) mice, as well as diminished capacity of
these receptor-decient animals to support stimulated mononu-
clear cell inltration into the CNS (Fife et al., 2000; Izikson et al.,
2000), might stem at least partially from the absence of CCR2
expression by the brain microvascular endothelium. Such a hy-
pothesis is supported by the nding that adoptive transfer of
myelin oligodendrocyte glycoprotein-sensitized T-cells from
wild-type mice exhibiting EAE to naive CCR2 (/) recipients
failed to produce disease symptomatology and yielded a dramat-
ically reduced mononuclear CNS inltrate (Fife et al., 2000).
Perhaps reecting a similar need for chemokine receptor expres-
sion by a tissue barrier, neutrophil transuroepithelial migration in
a model of urinary tract infection has been reported to be depen-
dent on epithelial expression of CXCR1, a receptor for the
chemokine IL-8 (Godaly et al., 2000). In vitro paradigms of
leukocyte transendothelial migration, wherein BMEC and leuko-
cytes from wild-type and receptor-decient mice are mixed and
matched, would be extremely valuable in resolving the specic
contribution of endothelial chemokine receptors to the extrava-
sation process at the BBB.
With the spectrum of chemokine targets in the CNS, including
all resident and many transient cell types, chemokine activity is
likely to affect homeostatic, developmental, and pathological pro-
cesses (Asensio and Campbell, 1999; Hesselgesser and Horuk,
1999; Bacon and Harrison, 2000). Although this discussion fo-
cused on the role of endothelial CCR2 in mediating leukocyte
extravasation, it well may be that brain microvascular expression
of this receptor also features prominently in the migration of
other cell types across the BBB during ontogeny and adulthood
Dzenko et al. CCR2 on Brain Microvessels J. Neurosci., December 1, 2001, 21(23):92149223 9221
(Rezaie and Male, 1999; Silverman et al., 2000). Endothelial
CCR2 additionally may play a role in angiogenesis in the brain, as
suggested by studies showing MCP-1-induced endothelial chemo-
taxis in vitro (Weber et al., 1999; Salcedo et al., 2000) and
formation of blood vessels in vivo (Salcedo et al., 2000). Accord-
ingly, the endothelium may prove to be an effective therapeutic
target for modifying CNS effects associated with aberrant MCP-1
activity.
REFERENCES
Abbott NJ, Romero IA (1996) Transporting therapeutics across the
bloodbrain barrier. Mol Med Today 2:106 113.
Amara A, Gall SL, Schwartz O, Salamero J, Montes M, Loetscher P,
Baggliolini M, Virelizier JL, Arenzana-Seisdedos F (1997) HIV co-
receptor downregulation as antiviral principle: SDF-1
-dependent in-
ternalization of the chemokine receptor CXCR4 contributes to inhibi-
tion of HIV replication. J Exp Med 186:139146.
Anderson RGW (1998) The caveolae membrane system. Annu Rev Bio-
chem 67:199 225.
Andjelkovic AV, Pachter JS (2000) Characterization of binding sites for
chemokines MC P-1 and M IP-1
on human brain microvessels. J Neu-
rochem 75:1898 1906.
Andjelkovic AV, Spencer DD, Pachter JS (1999) Visualization of che-
mokine binding sites on human brain microvessels. J Cell Biol
145:403412.
Andjelkovic AV, Zochowski M, Morgan F, Pachter JS (2001) Qualita-
tive and quantitative analysis of monocyte transendothelial migration
by confocal microscopy and three-dimensional image reconstruction. In
Vitro Cell Dev Biol Anim 37:111120.
Aragay AM, Mellado M, Frade JM, Martin AM, Jimenez-Sainz MC,
Martinez AC, Mayor Jr F (1998) Monocyte chemoattractant protein-
1-induced CCR2B receptor desensitization mediated by the G-protein-
coupled receptor kinase 2. Proc Natl Acad Sci USA 17:29852990.
Asensio VC, Campbell IL (1999) Chemokines in the CNS: plurifunc-
tional mediators in diverse states. Trends Neurosci 22:504512.
Bacon K B, Harrison JK (2000) Chemokines and their receptors in neu-
robiology: perspectives in physiology and homeostasis. J Neuroimmu-
nol 104:9297.
Banks WA, Kastin AJ (1991) Blood to brain transport of interleukin
links the immune and central nervous systems. Life Sci
48:PL117PL121.
Banks WA, Kastin AJ (1996) Reversible association of the cytokines
MI P-1
and MIP-1
with the endothelia of the bloodbrain barrier.
Neurosci Lett 205:202206.
Banks WA, Kastin AJ, Gutierrez EG (1994) Penetration of interleukin-6
across the bloodbrain barrier. Neurosci Lett 179:5356.
Benlimame N, Le PU, Nabi IR (1998) Localization of autocrine motility
factor receptor to caveolae and clathrin-independent internalization of
its ligand to smooth endoplasmic reticulum. Mol Biol Cell 9:17731786.
Berger O, Gan X, Gujuluva C, Burns AR, Suur G, Stins M, Way D, Witte
M, Weinand M, Said J, Kim K-S, Taub D, Graves MC, Fiala M (1999)
CXC and CC chemokine receptors on coronary and brain endothelia.
Mol Med 5:795805.
Berman JW, Guida MP, Warren J, Amat J, Brosnan CF (1996) Local-
ization of monocyte chemoattractant peptide-1 expression in the cen-
tral nervous system in experimental autoimmune encephalomyelitis
and trauma in the rat. J Immunol 156:30173023.
Biegel D, Spencer DD, Pachter JS (1995) Isolation and culture of human
brain microvessel endothelial cells: the study of blood-brain barrier
properties in vitro. Brain Res 692:183189.
Boddeke HWG, Zuurman M, Brouer N, Biber K (2000) Cloning and
characterization of a new chemokine receptor in mouse glial cells. Soc
Neurosci Abstr 26[part 2]:1944.
Bohm SK, Grady EF, Bunnett NW (1997) Regulatory mechanisms that
modulate signaling by G-protein-coupled receptors. Biochem J
322:118.
Dehouck B, Fenart L, Dehouck MP, Pierce A, Torpier G, Cecchelli RA
(1997) New function for the LDL receptor: transcytosis of LDL across
the bloodbrain barrier. J Cell Biol 138:877 889.
Deli MA, Descamps L, Dehouck M P, C ecchelli R, Joo F, Abraham C S,
Torpier G (1995) Exposure of tumor necrosis factor-
to luminal
membrane of bovine capillary endothelial cells cocultured with astro-
cytes induces a delayed increase of permeability and cytoplasmic stress
ber formation of actin. J Neurosci Res 41:717726.
Dobbie MS, Hurst RD, Klein NJ, Surtees RA (1999) Upregulation of
intercellular adhesion molecule-1 expression on human endothelial
cells by tumour necrosis factor alpha in an in vitro model of the
bloodbrain barrier. Brain Res 830:330336.
Edinger A, Mankowski J, Doranz BJ, Margulies BJ, Lee B, Rucker J,
Sharron M, Hoffman TL, Berson JF, Zink MC, Hirsch VM, Clements
JE, Doms RW (1997) CD4-independent, CCR5-dependent infection
of brain capillary endothelial cells by a neurovirulent simian immuno-
deciency virus strain. Proc Natl Acad Sci USA 94:1474214747.
Fantuzzi L, Borghi P, Ciolli V, Pavlakis G, Bellardelli F, Cessani S
(1999) L oss of CCR2 expression and functional response to monocyte
chemotactic protein (MCP-1) during the differentiation of human
monocytes: role of secreted MCP-1 in the regulation of the chemotactic
response. Blood 94:875883.
Feil C, Augustin HG (1998) Endothelial cells differently express func-
tional C XC-chemokine receptor-4 (CXCR-4/fusin) under the control
of autocrine activity and exogenous cytokines. Biochem Biophys Res
Commun 247:3845.
Feng D, Nagy JA, Pyne K , Hammel I, Dvorak HF, Dvorak AM (1999)
Pathways of macromolecular extravasation across microvascular endo-
thelium in response to VPF/VEGF and other vasoactive mediators.
Microcirculation 6:2344.
Feniger-Barish R, Ran M, Zaslaver A, Ben-Baruch A (1999) Differential
modes of regulation of CXC chemokine-induced internalization and
recycling of human CXCR1 and CXCR2. Cytokine 11:996 1009.
Fife BT, Huffnagle GB, Kuziel WA, Karpus WJ (2000) CC chemokine
receptor 2 is critical for induction of experimental autoimmune en-
cephalomyelitis. J Exp Med 192:899 905.
Friden PM (1993) Receptor-mediated transport of peptides and proteins
across the bloodbrain barrier. In: The bloodbrain barrier: cellular
and molecular biology (Pardridge WM, ed), pp 229 248. New York:
Raven.
Ghirnikar RS, Lee YL, Eng LF (1998) Inammation in traumatic brain
injury: role of cytokines and chemokines. Neurochem Res 23:329 340.
Glabinski AR, Balasingame V, Tani M, Kunkel SL, Streiter RM, Yong
VW, Ransohoff RM (1996) Chemokine monocyte chemoattractant
protein-1 is expressed by astrocytes after mechanical injury to the
brain. J Immunol 156:4363 4368.
Godaly G, Hang L, Frendeus B, Svanborg C (2000) Transepithelial neu-
trophil migration is CXCR1 dependent in vitro and is defective in IL -8
receptor knock-out mice. J Immunol 165:52875294.
Gupta SK, Lysko PG, Pillarisetti K, Ohlstein E, Stadel JM (1998) Che-
mokine receptors in human endothelial cells. J Biol Chem 273:4282
4287.
Gutierrez EG, Banks WA, Kastin AJ (1993) Murine tumor necrosis
factor alpha is transported from blood to brain in the mouse. J Neuro-
immunol 47:169 176.
Gutierrez EG, Banks WA, Kastin AJ (1994) Blood-borne interleukin-1
receptor antagonist crosses the bloodbrain barrier. J Neuroimmunol
55:153160.
Hesselgesser J, Horuk R (1999) Chemokines and chemokine receptors
in the brain. In: Chemokines in disease: biology and clinical research
(Hebert CA, ed), pp 295322. New Jersey: Humana.
Hoogewerf AJ, Kuschert GS, Proudfoot AE, Borlat F, Clark-Lewis I,
Power CA, Wells TN (1997) Glycosaminoglycans mediate cell surface
oligomerization of chemokines. Biochemistry 36:13670 13678.
Huang D, Han Y, Rani MR, Glabinski A, Trebst C, Sorensen T, Tani M,
Wang J, Chien P, OBryan S, Bielecki B, Zhou ZL, Majumder S,
Ransohoff RM (2000) Chemokines and chemokine receptors in in-
ammation of the nervous system: manifold roles and exquisite regu-
lation. Immunol Rev 177:52 67.
Ikezu T, Ueda H, Trapp BD, Nishiyama K, Sha JF, Volonte D, Galbiati
F, Byrd AL, Bassell G, Serizawa H, Lane WS, Lisanti MP, Okamoto T
(1998) Afnity-purication and characterization of caveolins from the
brain: differential expression of caveolin-1, -2, and -3 in brain endothe-
lial and astroglial cells. Brain Res 804:177192.
Isshiki M, Anderson RG (1999) Calcium signal transduction from caveo-
lae. Cell Calcium 26:201208.
Izikson L, Klein RS, Charo IF, Weiner HL, Luster AD (2000) Resis-
tance to experimental autoimmune encephalomyelitis in mice lacking
the CC chemokine receptor (CCR) 2. J Exp Med 192:10751080.
John TA, Vogel SM, Minshall RD, Ridge K, Tiruppathi C, Malik AB
(2001) Evidence for the role of alveolar epithelial gp60 in active
transalveolar albumin transport in the rat lung. J Physiol (Lond)
533:547559.
King GL, Johnson SM (1985) Receptor-mediated transport on insulin
across endothelial cells. Science 227:15831586.
Kniesel U, Wolburg H (2000) Tight junctions of the bloodbrain barrier.
Cell Mol Neurobiol 20:5776.
Kurzchalia TV, Parton RG (1999) Membrane microdomains and caveo-
lae. Curr Opin Cell Biol 11:424431.
Kuschert GS, Coulin F, Power CA, Proudfoot AE, Hubbard RE,
Hoogewerf AJ, Wells TN (1999) Glycosaminoglycans interact selec-
tively with chemokines and modulate receptor binding and cellular
responses. Biochemistry 38:12959 12968.
Kuziel WA, Morgan SJ, Dawson TC, Grifn S, Smithies O, Ley K, Maeda
N (1997) Severe reduction in leukocyte adhesion and monocyte ex-
travasation in mice decient in chemokine receptor 2. Proc Natl Acad
Sci USA 94:1205312058.
Lassmann H, Zimprich F, Vass K, Hickey WF (1991) Microglial cells
are a component of the perivascular glia limitans. J Neurosci Res
28:236243.
9222 J. Neurosci., December 1, 2001, 21(23):92149223 Dzenko et al. CCR2 on Brain Microvessels
Lefkowitz RJ (1998) G-protein-coupled receptors. III. New roles for
receptor kinases and
-arrestins in receptor signaling and desensitiza-
tion. J Biol Chem 273:1867718680.
Lisanti MP, Scherer PE, Vidugiriene J, Tang Z, Hermanowski-Vosatka
A, Tu YH, Cook RF, Sargiacomo M (1994) Characterization of
caveolin-rich membrane domains isolated from an endothelial-rich
source: implications for human disease. J Cell Biol 126:111126.
Lobie PE, Sadir R, Graichen R, Mertaini HC, Morel G (1999) Caveolar
internalization of growth hormone. Exp Cell Res 246:4755.
Mack M, Schlondorff D (2000) Downmodulation and recycling of che-
mokine receptors. Methods Mol Biol 138:191195.
Mack M, Luckow B, Nelson PJ, Cihak J, Simmons G, Clapham PR,
Signoret N, Marsh M, Stangassinger M, Borlat F, Wells TN, Schlon-
dorff D, Proudfoot AE (1998) Aminooxypentane-RANTES induces
CCR5 internalization but inhibits recycling: a novel inhibitory mecha-
nism of HIV infectivity. J Exp Med 187:12151224.
Madani N, Kozak SL, Kavanaugh MP, Kabat D (1998) gp120 envelope
glycoproteins of human immunodeciency viruses competitively antag-
onize signaling by coreceptors CXCR4 and CCR5. Proc Natl Acad Sci
USA 95:8005 8010.
Makic J, Stins M, McComb JG, Calero M, Ghiso J, K im KS, Yan DS,
Stern D, Schmidt AM, Frangione B, Zlokovic BV (1998) Human
bloodbrain barrier receptors for Alzheimers amyloid 1-40. J Clin
Invest 102:734 743.
Megyeri P, Abraham C S, Tmesvari P, Kovacs J, Vas T, Speer C P (1992)
Recombinant human tumor necrosis factor alpha constricts pial arte-
rioles and increases bloodbrain barrier permeability in newborn pig-
lets. Neurosci Lett 148:137140.
Middleton J, Neil S, Wintle J, Clark-Lewis I, Moore H, Lam C, Auer M,
Hub E, Rot A (1997) Transcytosis and surface presentation of IL-8 by
venular endothelial cells. Cell 91:385395.
Miller RJ, Meucci O (1999) AIDS and the brain: is there a chemokine
connection? Trends Neurosci 22:471 479.
Molino M, Woolkalis MJ, Prevost N, Pratico D, Barnathan ES, Tarabo-
letti G, Haggart BC, Hesselgesser J, Horuk R, Hoxie JA, Brass LF
(2000) C XCR4 on human endothelial cells can serve as both a medi-
ator of biological responses and as a receptor for HIV-2. Biochim
Biophys Acta 1500:227240.
Murdoch C, Monk PN, Finn A (1999) CXC chemokine receptor expres-
sion on human endothelial cells. Cytokine 11:704 712.
Okamoto T, Schlegel A, Scherer PE, Lisanti MP (1998) C aveolins, a
family of scaffolding proteins for organizing preassembled signaling
complexes at the plasma membrane. J Biol Chem 273:5419 5422.
Okamoto Y, Ninomiya H, Miwa S, Masaski T (2000) Cholesterol oxida-
tion switches the internalization pathway of endothelin receptor A from
caveolae to clathrin-coated pits in Chinese hamster ovary cells. J Biol
Chem 275:64396446.
Orlandi PA, Fishman PH (1998) Filipin-dependent inhibition of cholera-
toxin: evidence for toxin internalization and activation through
caveolae-like domains. J Cell Biol 141:905915.
Ransohoff RM, Hamilton TA, Tani M, Stoler M, Shick HE, Major JA,
Estes ML, Thomas DM, Tuohy VK (1993) Astrocyte expression of
mRNA encoding cytokines IP-10 and JE/MCP-1 in experimental
autoimmune encephalomyelitis. FASEB J 7:592600.
Rezaie P, Male D (1999) Colonisation of the developing human brain
and spinal cord by microglia: a review. Microsc Res Tech 45:359382.
Roettger BF, Rentsch RU, Pinon D, Holicky E, Hadac E, Larkin JM,
Miller L J (1995) Dual pathways of internalization of the cholecysto-
kinin receptor. J Cell Biol 128:1029 1041.
Rose JM, Audus KL (1998) Receptor-mediated transport of angiotensin
by brain microvessel endothelial cells. Peptides 19:10231030.
Roseberry AG, Hosey MM (2001) Internalization of the M
2
muscarinic
acetylcholinergic receptor proceeds through an atypical pathway in
HEK293 cells that is independent of clathrin and caveolae. J Cell Sci
114:739746.
Rossi D, Zlotnick A (2000) The biology of chemokines and chemokine
receptors. Annu Rev Immunol 18:217242.
Rothberg K, Heuser JE, Donzell WC, Ying Y-S, Glenney JR, Anderson
RGW (1992) Caveolin, a protein component of caveolae membrane
coats. Cell 68:673 682.
Rottman JB, Ganley KP, Williams K, Wu L, Mackay CR, Ringler DJ
(1997) C ellular localization of the chemokine receptor CCR5. Corre-
lation to cellular targets of HIV-1 infection. Am J Pathol 15:13411351.
Rubin LL, Staddon JM (1999) The cell biology of the bloodbrain
barrier. Annu Rev Neurosci 22:1128.
Salcedo R, Ponce ML, Young HA, Wasserman K, Kleinman HK, Op-
penheim JJ, Murphy WJ (2000) Human endothelial cells express
CCR2 and respond to MCP-1: direct role of MCP-1 angiogenesis and
tumor progression. Blood 96:34 40.
Sanders VJ, Pittman CA, Whit MG, Wiley CA, Achim CL (1998) Che-
mokines and receptors in HIV encephalitis. AIDS 12:10211026.
Sarau HM, Rush JA, Foley JJ, Brawner ME, Schmidt DB, White JR,
Barnette MS (1997) Characterization of functional chemokine recep-
tors (CCR1 and CCR2) on EoL-3 cells: a model system to examine the
role of chemokines in cell f unction. J Pharmacol Exp Ther 283:411
418.
Sauder C, Hallensleben W, Pagenstecher A, Schneckenburger S, Biro L,
Pertlik D, Hausmann J, Suter M, Staeheli P (2000) Chemokine gene
expression in astrocytes of Borna disease virus-infected rats and mice
in the absence of inammation. J Virol 74:92679380.
Schlegel A, Lisanti MP (2001) Caveolae and their coat proteins, the
caveolins: from electron microscopic novelty to biological launching
pad. J Cell Physiol 186:329 337.
Schnitzer JE, Oh P, Pinney E, Allard J (1994) Filipin-sensitive caveolae-
mediated transport in endothelium: reduced transcytosis, scavenger
endocytosis, and capillary permeability of select macromolecules.
J Cell Biol 127:12171232.
Schraufstatter IU, Chung J, Burger M (2001) IL -8 activates endothelial
cell CXCR1 and CXCR2 through Rho and Rac signaling pathways.
Am J Physiol Lung Cell Mol Physiol 280:L1094 L1103.
Sharafeldin A, Eltayeb R, Pashenkov M, Bakhiet M (2000) Chemokines
are produced in the brain early during the course of experimental
African trypanosomiasis. J Neuroimmunol 103:165170.
Shaul RW, Anderson RG (1998) Role of plasmalemma caveolae in sig-
nal transduction. Am J Physiol 275:L843L851.
Shaw KTY, Grieg NH (1999) Chemokine receptor mRNA expression at
the in vitro blood-brain barrier during HIV infection. NeuroReport
10:5356.
Silverman A-J, Sutherland AK , Wilhelm M, Silver R (2000) Mast cells
migrate from blood to brain. J Neurosci 20:401 408.
Van Der Voorn P, Tekstra J, Beelen RH, Tensen CP, Van Der Walk P,
De Groot CJ (1999) Expression of MC P-1 by reactive astrocytes in
demyelinating multiple sclerosis lesions. Am J Pathol 154:4551.
Volin MV, Joseph L, Shockley MS, Davis PF (1998) Chemokine recep-
tor CXCR4 expression in endothelium. Biochem Biophys Res Com-
mun 242:46 53.
Vorbrodt AW, Dobrowgowska DH, Lossinsky AS, Wisniewski HM
(1994) Ultrastructural localization of lectin receptors on the luminal
and abluminal aspects of brain micro-blood vessels. J Histochem C y-
tochem 34:251261.
Weber KS, Nelson NJ, Grone HJ, Weber C (1999) E xpression of CCR2
by endothelial cells: implications for MCP-1-mediated wound injury
repair and in vivo inammatory activation of endothelium. Arterioscler
Thromb Vasc Biol 19:20852093.
Wong D, Prameya R, Dorivini-Zis K (1999) In vitro adhesion and mi-
gration of T-lymphocytes across monolayers of human brain microves-
sel endothelial cells: regulation by ICAM-1, VCAM-1, E-selectin, and
PECAM-1. J Neuropathol Exp Neurol 58:138 152.
Yang W, Wang D, Richmond A (1999) Role of clathrin-mediated endo-
cytosis in CXCR2 sequestration, resensitization, and signal transduc-
tion. J Cell Biol 274:11328 11333.
Dzenko et al. CCR2 on Brain Microvessels J. Neurosci., December 1, 2001, 21(23):92149223 9223
... Indeed, it has been suggested that upon chemokine binding, ACKR1 translocates intracellularly, where it colocalizes with caveolin in intracellular vesicles in Madin-Darby canine kidney (MDCK) cells [43], suggesting caveolae as an intracellular mechanism of transport of ACKR1. In addition, it has been shown that CCL2 can be transported from the abluminal to the luminal side of brain microvascular endothelial cells by a transcellular mechanism that at least to a certain degree involves binding of CCL2 to CCR2 and caveolae [45,46]. Our pMBMECs constitutively express caveolin-1 at the mRNA and protein level, with caveolin-1 expression however not further upregulated upon cytokine stimulation of the pMB-MECs. ...
Article
Full-text available
The migration of CD4⁺ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in multiple sclerosis or its animal model, experimental autoimmune encephalomyelitis (EAE). T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data supports that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation. This article is protected by copyright. All rights reserved
... Key chemokines secreted by monocytes, including interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1), which regulate firm monocyte adhesion and extravasation from the vascular endothelium, have been identified by molecular binding assays. 22,23 Electrophoresis, microarrays, and polymerase chain reaction (PCR) techniques have allowed for the evaluation of monocyte and endothelial gene and protein expression that correlate with extravasation events in response to inflammatory factors. For example, lipopolysaccharides (LPS) induce E-selectin, VCAM-1, and ICAM-1 gene expressions, 24 whereas interferon gamma (INF-γ) and interleukin-4 (IL-4) upregulate MCP-1 mRNA and protein expressions. ...
Article
Full-text available
Extravasation of circulating cells is an essential process that governs tissue inflammation and the body's response to pathogenic infection. To initiate anti-inflammatory and phagocytic functions within tissues, immune cells must cross the vascular endothelial barrier from the vessel lumen to the subluminal extracellular matrix. In this work, we present a microfluidic approach that enables the recreation of a three-dimensional, perfused endothelial vessel formed by human endothelial cells embedded within a collagen-rich matrix. Monocytes are introduced into the vessel perfusate, and we investigate the role of luminal flow and collagen concentration on extravasation. In vessels conditioned with the flow, increased monocyte adhesion to the vascular wall was observed, though fewer monocytes extravasated to the collagen hydrogel. Our results suggest that the lower rates of extravasation are due to the increased vessel integrity and reduced permeability of the endothelial monolayer. We further demonstrate that vascular permeability is a function of collagen hydrogel mass concentration, with increased collagen concentrations leading to elevated vascular permeability and increased extravasation. Collectively, our results demonstrate that extravasation of monocytes is highly regulated by the structural integrity of the endothelial monolayer. The microfluidic approach developed here allows for the dissection of the relative contributions of these cues to further understand the key governing processes that regulate circulating cell extravasation and inflammation.
... CCL2 binds to the C-C motif chemokine receptor 2 (CCR2), which is expressed by neurons (18), human fetal astrocytes (19) and brain microvascular endothelial cells (BMECs) (20). CCL2 also binds to the D6 chemokine decoy receptor, which is expressed on adult human astrocytes (21). ...
Article
Full-text available
C-C motif chemokine ligand 2 (CCL2) is a chemoattractant for leukocytes including monocytes, T cells, and natural killer cells and it plays an important role in maintaining the integrity and function of the brain. However, there is accumulating evidence that many neurological diseases are attributable to a dysregulation of CCL2 expression. Acquired immune deficiency syndrome (AIDS) encephalopathy is a severe and frequent complication in individuals infected with the human immunodeficiency virus (HIV) or the simian immunodeficiency virus (SIV). The HIV and SIV Nef protein, a progression factor in AIDS pathology, can be transferred by microvesicles including exosomes and tunneling nanotubes (TNT) within the host even to uninfected cells, and Nef can induce CCL2 expression. This review focuses on findings which collectively add new insights on how Nef-induced CCL2 expression contributes to neurotropism and neurovirulence of HIV and SIV and elucidates why adjuvant targeting of CCL2 could be a therapeutic option for HIV-infected persons.
... As the effects of DAG kinase inhibition and DAG lipase inhibition are mimicked by OAG and not rescued by addition of exogenous PIP 2 , the effect on CCL2 signalling of inhibiting DAG metabolism is likely to be due to the accumulation of DAG rather than limitation of a DAG metabolite or PIP 2 synthesis.If this is the case, the data suggest that there is no redundancy between kinase-or lipase-dependent metabolic pathways for DAG in human monocytes and processes that modulate the activity of these enzymes have the potential to modulate CCL2 signalling efficacy. Although CCR2 receptors are classical GPCRs and can undergo internalisation as part of mechanisms of receptor desensitisation(Dzenko, Andjelkovic, Kuziel, & Pachter, 2001;Volpe et al., 2012), the data presented here suggest that the mechanism via which inhibition of DAG metabolism causes loss of CCL2 signalling efficacy is not via a reduction in the cell surface population of CCR2 receptors. This therefore suggests that the inhibitory mechanism must involve modulation of the CCR2 receptor itself, an auxiliary subunit or a downstream effector protein.As the effects of OAG, DAG kinase inhibition, and DAG lipase inhibition are reversed by the broadspectrum PKC inhibitor Gö6983 but not Gö6976, a selective inhibitor of Ca 2+ -dependent PKC isoforms, this suggests that the inhibitor effects of DAG accumulation are mediated by a Ca 2+ -independent PKC isoform. ...
Article
Full-text available
Background and Purpose CCL2 is an inflammatory chemokine that stimulates the recruitment of monocytes into tissue via activation of the GPCR CCR2. Experimental Approach Freshly isolated human monocytes and THP‐1 cells were used. Fura‐2 loaded cells were used to measure intracellular Ca²⁺ responses. Transwell migration to measure chemotaxis. siRNA‐mediated gene knock‐down was used to support pharmacological approaches. Key Results CCL2 evoked intracellular Ca²⁺ signals and stimulated migration in THP‐1 monocytic cells and human CD14⁺ monocytes in a CCR2‐dependent fashion. Attenuation of DAG catabolism in monocytes by inhibiting DAG kinase (R59949) or DAG lipase (RHC80267) activity suppressed CCL2‐evoked Ca²⁺ signalling and transwell migration in monocytes. These effects were not due to a reduction in the number of cell surface CCR2. The effect of inhibiting DAG kinase or DAG lipase could be mimicked by addition of the DAG analogue 1‐oleoyl‐2‐acetyl‐sn‐glycerol (OAG) but was not rescued by application of exogenous phosphatidylinositol 4,5‐bisphosphate. Suppressive effects of R59949, RHC80267, and OAG were partially or fully reversed by Gö6983 (pan PKC isoenzyme inhibitor) but not by Gö6976 (PKCα and PKCβ inhibitor). RNAi‐mediated knock‐down of DAG kinase α isoenzyme modulated CCL2‐evoked Ca²⁺ responses in THP‐1 cells. Conclusions and Implications Taken together, these data suggest that DAG production resulting from CCR2 activation is metabolised by both DAG kinase and DAG lipase pathways in monocytes and that pharmacological inhibition of DAG catabolism or application suppresses signalling on the CCL2–CCR2 axis via a mechanism dependent upon a PKC isoenzyme that is sensitive to Gö6983 but not Gö6976.
... Quantita- tive region of interest (ROI) analysis of immunostained histolog- ical sections showed no evidence for increased albumin or IgG extravasation, and we did not observe focal areas of increased staining, suggesting ''normal'' BBB permeability ( Figure S3C). Since multiple studies suggest that monocyte infiltration into the diseased or injured CNS is facilitated by CCR2 (Ajami et al., 2011;Dzenko et al., 2001), we wondered whether engraftment of MLCs was similarly CCR2 dependent in Csf1r À/À hosts. We found that, as with WT BM, i.p. injection of CCR2 Rfp/Rfp (Ccr2 knockout) BM (Saederup et al., 2010) into Csf1r À/À mice leads to robust TMEM119+ MLC engraftment at 2 weeks, meaning that CCR2 is dispensable in this system ( Figure 3E). ...
Article
Microglia, the brain's resident macrophages, are dynamic CNS custodians with surprising origins in the extra-embryonic yolk sac. The consequences of their distinct ontogeny are unknown but critical to understanding and treating brain diseases. We created a brain macrophage transplantation system to disentangle how environment and ontogeny specify microglial identity. We find that donor cells extensively engraft in the CNS of microglia-deficient mice, and even after exposure to a cell culture environment, microglia fully regain their identity when returned to the CNS. Though transplanted macrophages from multiple tissues can express microglial genes in the brain, only those of yolk-sac origin fully attain microglial identity. Transplanted macrophages of inappropriate origin, including primary human cells in a humanized host, express disease-associated genes and specific ontogeny markers. Through brain macrophage transplantation, we discover new principles of microglial identity that have broad applications to the study of disease and development of myeloid cell therapies. Bennett et al. create a macrophage transplantation system to measure how origin and brain environment contribute to microglial identity. Although diverse macrophage types survive in the brain, only those sharing developmental origins with microglia express microglial genes normally.
Chapter
Interactions between the immune and nervous systems are involved in many disease processes. Modulation of inflammation can provide an important opportunity to enhance regeneration within the central nervous system. This authoritative book defines the key cellular players in mounting an inflammatory response and highlights critical factors in the target organ that influence the nature of that response and its capacity either to damage or protect the brain. Several key clinical areas are highlighted – particularly autoimmune diseases of the nervous system including multiple sclerosis, as well as microbiological and traumatic challenges; the book therefore provides both a summary of the basic science background as well as practical, clinical-friendly guidelines to management. The book will be of interest to a wide range of physicians, including neurologists, neurosurgeons, neurorehabilitationists, infectious disease physicians, and clinical neuroscientists, as well as neuroscientists and immunologists.
Article
The brain microvasculature is altered in normal aging and in the presence of disease processes, such as neurodegeneration or ischemia, but there are few methods for studying living tissues. We now report that viable microvessels (MV) are readily isolated from brain tissue of subjects enrolled in studies of neurodegenerative diseases who undergo rapid autopsy (performed with <12 h postmortem interval – PMI). We find that these MV retain their morphology and cellular components and are fairly uniform in size. Sufficient MV (~3–5000) is obtained from 3 to 4 g of tissue to allow for studies of cellular composition as well as ECM. Using live/dead assays, these MV are viable for up to 5 days in tissue culture media (2D) designed to support endothelial cells and up to 11 days post-isolation in a 3-dimensional (3D) matrix (Low Growth Factor Matrigel™). Assays that measure the reducing potential of live cells, and thus metabolism, demonstrated that the majority of the MV maintain high levels of activity for a similar number of days in vitro as the viability studies. Functional cellular components (such as tight junctions and transporter proteins) and extracellular matrix (ECM) of MV in tissue culture media, and to a lesser extent in 3D matrices, were readily visualized using immunofluorescence techniques. MV in tissue culture media are lysed and protein content analyzed, but MV in 3D matrix first require removal of the supporting matrix, which can confound the analysis of MV ECM. Finally, MV can be preserved in cryoprotective media, whereby over 50% retain their baseline viability upon thawing. In summary, we find that MV isolated from human brains undergoing rapid autopsy are viable in standard tissue culture for up to 5 days and the timeframe for experiments can be extended up to 11 days by use of a supportive 3D matrix. Viable human MV allow for temporal and spatial analysis of relevant cellular and ECM components that have implications for microvascular function in neurodegenerative diseases, vascular brain injury, and neurotrauma.
Article
Objective: Chemokines regulate infiltration of immune cells to brain in inflammation. Cathepsin C (CatC), a lysosomal protease, has been found to participate in neuroinflammation. However, how CatC affects chemokines expression in neuroinflammation triggered by traumatic brain injury (TBI) remains unclear. Here, we investigated the effects of CatC on chemokines and neuroinflammation in TBI. Methods: The present study used CatC knockdown (KD) and overexpression (OE) mice to generate cryogenic brain lesion model and determined effects of CatC on expression of chemokines CCL2, CCL5 and CXCL2 and infiltration of immune cells in acute and chronic phases of the lesion. Further, cellular sources of various chemokines were demonstrated in vitro. Values were compared with wild type (WT) mice. Results: The results found that 6 h after lesion, CatC expression,IL-1β and TNF-α mRNA and protein expression were strongly induced in the lesions; CCL2 and CXCL2 mRNA and protein expression were increased in CatC OE mice, while decreased in CatC KD mice. On the 3rd day after lesion, macrophages and neutrophils were mainly infiltrated to the lesions. Simultaneously, Iba-1+ cells in CatC OE mice were increased, while MPO + cells in CatC KD mice were decreased. In contrast, on the 28th day after lesion, a few lymphocytes were infiltrated surrounding new blood vessels. CatC OE mice showed larger volumes of scar areas, higher expression of CCL2,CXCL2,IL-1β,TNF-α,IL-6 and iNOS, as well as stronger GFAP+ and Iba-1+ signals, while CatC KD mice had reversed effects. No significant differences of CCL5 expression were found in various genotype mice. Further, in vitro study demonstrated CatC-induced expression of CCL2 were mainly derived from microglia and neurons, while CXCL2 derived from microglia and astrocytes. Conclusion: Our data indicate that CatC aggravates neuroinflammation via promoting production of CCL2 and CXCL2 in glial cells and neurons in a cryogenic brain lesion, providing potential cellular and molecular targets for future intervention of TBI and other neuroinflammatory diseases.
Article
In atherosclerosis, resident vascular smooth muscle cells (VSMCs) in the blood vessels become highly plastic and undergo phenotypic switching from the quiescent, contractile phenotype to the migratory and proliferative, synthetic phenotype. Additionally, recent VSMC lineage-tracing mouse models of atherosclerosis have found that VSMCs transdifferentiate into macrophage-like and osteochondrogenic cells and make up to 70% of cells found in atherosclerotic plaques. Given VSMC phenotypic switching is regulated by microRNA-145 (miR-145), we hypothesized that nanoparticle-mediated delivery of miR-145 to VSMCs has the potential to mitigate atherosclerosis development by inhibiting plaque-propagating cell types derived from VSMCs. To test our hypothesis, we synthesized miR-145 micelles targeting the C-C chemokine receptor-2 (CCR2), which is highly expressed on synthetic VSMCs. When miR-145 micelles were incubated with human aortic VSMCs in vitro, >90% miR-145 micelles escaped the lysosomal pathway in 4 hours and released the miR cargo under cytosolic levels of glutathione, an endogenous reducing agent. As such, miR-145 micelles rescued atheroprotective contractile markers, myocardin, α-SMA, and calponin, in synthetic VSMCs in vitro. In early-stage atherosclerotic ApoE-/- mice, one dose of miR-145 micelles prevented lesion growth by 49% and sustained an increased level of miR-145 expression after 2 weeks post-treatment. Additionally, miR-145 micelles inhibited 35% and 43% plaque growth compared to free miR-145 and PBS, respectively, in mid-stage atherosclerotic ApoE-/- mice. Collectively, we present a novel therapeutic strategy and cell target for atherosclerosis, and present miR-145 micelles as a viable nanotherapeutic that can intervene atherosclerosis progression at both early and later stages of disease.
Article
Experimental autoimmune encephalomyelitis is a CD4+ T cell-mediated demyelinating disease of the CNS that serves as a model for multiple sclerosis. Cytokines and chemokines shape Th1 and Th17 effector responses as well as regulate migration of leukocytes to the CNS during disease. The CNS cellular infiltrate consists of Ag-specific and nonspecific CD4+ and CD8+ T cells, neutrophils, B cells, monocytes, macrophages, and dendritic cells. The mechanism of immune-mediated inflammation in experimental autoimmune encephalomyelitis has been extensively studied in an effort to develop therapeutic modalities for multiple sclerosis and, indeed, has provided insight in modern drug discovery. The present Brief Review highlights critical pathogenic aspects of cytokines and chemokines involved in generation of effector T cell responses and migration of inflammatory cells to the CNS. Select cytokines and chemokines are certainly important in the regulatory response, which involves T regulatory, B regulatory, and myeloid-derived suppressor cells. However, that discussion is beyond the scope of this brief review.
Article
Full-text available
Chemokines play an important role in the regulation of endothelial cell (EC) function, including proliferation, migration and differentiation during angiogenesis, and re-endothelialization after injury. In this study, reverse transcriptase-polymerase chain reaction was used to reveal expression of various CXC and CC chemokine receptors in human umbilical vein EC. Northern analysis showed that CXCR4 was selectively expressed in vascular EC, but not in smooth muscle cells. Compared with other chemokines, stromal cell-derived factor-1α (SDF-1α), the known CXCR4 ligand, was an efficacious chemoattractant for EC, causing the migration of ∼40% input cells with an EC50 of 10–20 nm. Of the chemokines tested, only SDF-1α induced a rapid, though variable mobilization of intracellular Ca2+in EC. Experiments with actinomycin D demonstrated that CXCR4 transcripts were short-lived, indicating a rapid mRNA turnover. Interferon-γ (IFN-γ) caused a pronounced down-regulation of CXCR4 mRNA in a concentration- and time-dependent manner. In a striking functional correlation, IFN-γ treatment also attenuated the chemotactic response of EC to SDF-1α. IL-1β, tumor necrosis factor-α, and lipopolysaccharide produced a time course-dependent biphasic effect on CXCR4 transcription. Expression of CXCR4 in EC is significant, more so as it and several CC chemokine receptors have been shown to serve as fusion co-receptors along with CD4 during human immunodeficiency virus infection. Taken together, these findings provide evidence of chemokine receptor expression in EC and offer an explanation for the action of chemokines like SDF-1α on the vascular endothelium.
Article
By 24 h after mechanical trauma to the cerebral cortex, astroglial reaction begins and injury sites are infiltrated by activated mononuclear phagocytes derived from blood-borne monocytes and endogenous microglia. There is little information about cellular interactions between astrocytes and leukocytes during this process. We previously showed that murine astrocytes produce chemokines including monocyte chemoattractant protein-1 (MCP-1) during experimental autoimmune encephalomyelitis. In this study, we asked whether astrocytes produce MCP-1 in the absence of immune mediated inflammation. To address this question, we analyzed the time course and cellular source of MCP-1 in mouse brain after penetrating mechanical injury, with particular focus on early time points before histologic detection of infiltrating mononuclear phagocytes. We observed sharply increased steady state levels of MCP-1 mRNA within 3 h after nitrocellulose membrane stab or implant injury to the adult mouse brain, and MCP-1 protein elevations were documented at 12 h postinjury. In situ hybridization combined with immunohistochemistry for the glial fibrillary acidic protein astrocyte marker showed that astrocytes were the cellular source of MCP-1 mRNA at these early time points after mechanical brain injury. Stab injury to the neonatal brain evoked neither MCP-1 expression nor astrogliosis. These results demonstrate that chemokine gene expression comprises one component of the astrocyte activation program. The data are consistent with a role for MCP-1 in the central nervous system inflammatory response to trauma.
Article
Although several CXC chemokines have been shown to induce angiogenesis and play roles in tumor growth, to date, no member of the CC chemokine family has been reported to play a direct role in angiogenesis. Here we report that the CC chemokine, monocyte chemotactic protein 1 (MCP-1), induced chemotaxis of human endothelial cells at nanomolar concentrations. This chemotactic response was inhibited by a monoclonal antibody to MCP-1. MCP-1 also induced the formation of blood vessels in vivo as assessed by the chick chorioallantoic membrane and the matrigel plug assays. As expected, the angiogenic response induced by MCP-1 was accompanied by an inflammatory response. With the use of a rat aortic sprouting assay in the absence of leukocytic infiltrates, we ruled out the possibility that the angiogenic effect of MCP-1 depended on leukocyte products. Moreover, the direct effect of MCP-1 on angiogenesis was consistent with the expression of CCR2, the receptor for MCP-1, on endothelial cells. Assessment of supernatant from a human breast carcinoma cell line demonstrated the production of MCP-1. Treatment of immunodeficient mice bearing human breast carcinoma cells with a neutralizing antibody to MCP-1 resulted in significant increases in survival and inhibition of the growth of lung micrometastases. Taken together, our data indicate that MCP-1 can act as a direct mediator of angiogenesis. As a chemokine that is abundantly produced by some tumors, it can also directly contribute to tumor progression. Therefore, therapy employing antagonists of MCP-1 in combination with other inhibitors of angiogenesis may achieve more comprehensive inhibition of tumor growth.
Article
Caveolae are specialized plasmalemmal microdomains originally studied in numerous cell types for their involvement in the transcytosis of macromolecules. They are enriched in glycosphingolipids, cholesterol, sphingomyelin, and lipid-anchored membrane proteins, and they are characterized by a light buoyant density and resistance to solubilization by Triton X-100 at 4°C. Once the identification of the marker protein caveolin made it possible to purify this specialized membrane domain, it was discovered that caveolae also contain a variety of signal transduction molecules. This includes G protein-coupled receptors, G proteins and adenylyl cyclase, molecules involved in the regulation of intracellular calcium homeostasis, and their effectors including the endothelial isoform of nitric oxide synthase, multiple components of the tyrosine kinase-mitogen-activated protein kinase pathway, and numerous lipid signaling molecules. More recent work has indicated that caveolae further serve to compartmentalize, modulate, and integrate signaling events at the cell surface. This specialized plasmalemmal domain warrants direct consideration in future investigations of both normal and pathological signal transduction in pulmonary cell types.
Chapter
Cytokines play a major role in orchestrating the response of the immune system to invasion by pathogenic organisms. In the late 1980s several cytokines were described that not only modulated the activity of immune cells but also elicited their migratory responses. These newly described cytokines, of which interleukin 8 (IL-8) was a prototypic member, delineated a new branch of cytokines called chemokines (chemo-tactic cytokines). Chemokines are small mainly basic proteins that have been classified into four distinct families, CXC, CC, C, and CX3C, based on the position of their con-served cysteine residues. The two major branches are the CXC and CC chemokines. CXC chemokines include IL-8 and melanoma growth stimulatory activity (MGSA) and are mainly involved in acute inflammation, inducing the migration of neutrophils. The CC chemokines include RANTES, MCP-1 and MIP-1α and are primarily involved in chronic inflammation chemoattracting monocytes and T cells.
Article
Receptor molecules play a major role in the desensitization of agonist-stimulated cellular responses. For G protein-coupled receptors, rapid desensitization occurs via receptor phosphorylation, sequestration, and internalization, yet the cellular compartments in which these events occur and their interrelationships are unclear. In this work, we focus on the cholecystokinin (CCK) receptor, which has been well characterized with respect to phosphorylation. We have used novel fluorescent and electron-dense CCK receptor ligands and an antibody to probe receptor localization in a CCK receptor-bearing CHO cell line. In the unstimulated state, receptors were diffusely distributed over the plasmalemma. Agonist occupation stimulated endocytosis via both clathrin-dependent and independent pathways. The former was predominant, leading to endosomal and lysosomal compartments, as well as recycling to the plasmalemma. The clathrin-independent processes led to a smooth vesicular compartment adjacent to the plasmalemma resembling caveolae, which did not transport ligand deeper within the cell. Potassium depletion largely eliminated clathrin-dependent endocytosis, while not interfering with agonist-stimulated receptor movement into subplasmalemmal smooth vesicle compartments. These cellular endocytic events can be related to the established cycle of CCK receptor phosphorylation and dephosphorylation, which we have previously described (Klueppelberg, U. G., L. K. Gates, F. S. Gorelick, and L. J. Miller. 1991. J. Biol. Chem. 266:2403-2408; Lutz, M. P., D. I. Pinon, L. K. Gates, S. Shenolikar, and L. J. Miller. 1993. J. Biol. Chem. 268:12136-12142). The rapid onset and peak of receptor phosphorylation after agonist occupation correlates best with a plasmalemmal localization, while stimulated receptor phosphatase activity correlates best with receptor residence in intracellular compartments. We postulate that the smooth vesicular compartment adjacent to the plasmalemma functions for the rapid resensitization of the receptor, while the classical clathrin-mediated endocytotic pathway is key for receptor downregulation via lysosomal degradation, as well as less rapid resensitization.
Article
Has there ever been a more confusing field in biology with respect to nomenclature than that of chemokines? Nomenclature for cytokines has been extremely difficult to control and the chemokine gene superfamily, whose members exhibit a bewildering array of names, is no exception. There are no consistent naming conventions within the superfamily and most chemokines have multiple names. Despite this, the first section of this book contains an attempt to place the nomenclature issue in perspective. Recent advances in genome sequencing have caused a disproportionate increase in our knowledge of a number of gene families and the chemokines are no exception. This is covered quite well in the first section of the book, in which tables listing the various chemokines and their receptors are presented. In addition, because the other area in which major advances in chemokine biology are being made is in the use of animal models, the last two chapters in this section are devoted to recent information arising from the use of genetically modified mice. The results of experiments on mice rendered genetically deficient in several chemokines and chemokine receptors is summarized by Craig Gerard and the results of a large body of work using mice transgenic or deficient in one of the prototype chemokines, MCP-1, is summarized with great care by Barrett Rollins and colleagues.