ArticlePDF Available

CXC and CC Chemokine Receptors on Coronary and Brain Endothelia

Authors:

Abstract and Figures

Chemokine receptors on leukocytes play a key role in inflammation and HIV-1 infection. Chemokine receptors on endothelia may serve an important role in HIV-1 tissue invasion and angiogenesis. The expression of chemokine receptors in human brain microvascular endothelial cells (BMVEC) and coronary artery endothelial cells (CAEC) in vitro and cryostat sections of the heart tissue was determined by light and confocal microscopy and flow cytometry with monoclonal antibodies. Chemotaxis of endothelia by CC chemokines was evaluated in a transmigration assay. In BMVEC, the chemokine receptors CCR3 and CXCR4 showed the strongest expression. CXCR4 was localized by confocal microscopy to both the cytoplasm and the plasma membrane of BMVEC. In CAEC, CXCR4 demonstrated a strong expression with predominantly periplasmic localization. CCR5 expression was detected both in BMVEC and CAEC but at a lower level. Human umbilical cord endothelial cells (HUVEC) expressed strongly CXCR4 but only weakly CCR3 and CCR5. Two additional CC chemokines, CCR2A and CCR4, were detected in BMVEC and CAEC by immunostaining. Immunocytochemistry of the heart tissues with monoclonal antibodies revealed a high expression of CXCR4 and CCR2A and a low expression of CCR3 and CCR5 on coronary vessel endothelia. Coronary endothelia showed in vitro a strong chemotactic response to the CC chemokines RANTES, MIP-1alpha, and MIP-1beta. The endothelia isolated from the brain display strongly both the CCR3 and CXCR4 HIV-1 coreceptors, whereas the coronary endothelia express strongly only the CXCR4 coreceptor. CCR5 is expressed at a lower level in both endothelia. The differential display of CCR3 on the brain and coronary endothelia could be significant with respect to the differential susceptibility of the heart and the brain to HIV-1 invasion. In addition, CCR2A is strongly expressed in the heart endothelium. All of the above chemokine receptors could play a role in endothelial migration and repair.
Content may be subject to copyright.
Molecular
Medicine
5:
795-805,
1999
Molecular
Medicine
0
1999
The
Picower
Institute
Press
CXC
and
CC
Chemokme
Receptors
on
Coronary
and
Brain
Endothelia
Omri
Berger,"8
Xiaohu
Gan,2'8
Chandrasekhar
Gujuluva,"
8
Alan
R.
Burns,4
Girija
Sulur,3
Monique
Stins,'
Dennis
Way,6
Marlys
Witte,6
Martin
Weinand,6
Jonathan
Said,3
Kwang-Sik
Kim,'
Dennis
Taub,7
Michael
C.
Graves,'
and
Milan
Fiala8
Departments
of
'Neurology,
2Microbiology
and
Immunology,
and
3Pathology,
UCLA
School
of
Medicine,
Los
Angeles,
California,
U.S.A.
4Department
of
Medicine,
Section
of
Cardiovascular
Sciences
and
Leukocyte
Biology,
Baylor
College
of
Medicine,
Houston,
Texas,
U.S.A.
5Division
of
Infectious
Diseases,
Children's
Hospital
of
Los
Angeles,
Los
Angeles,
California,
U.S.A.
6Department
of
Surgery,
University
of
Arizona,
Tucson,
Arizona,
U.S.A.
7National
Institute
on
Aging,
Baltimore,
Maryland,
U.S.A.
8Department
of
Medicine,
West
Los
Angeles
VA
Medical
Center,
Los
Angeles,
California,
U.S.A.
Accepted
October
25,
1999.
Abstract
Background:
Chemokine
receptors
on
leukocytes
play
a
key
role
in
inflammation
and
HIV-
1
infection.
Chemo-
kine
receptors
on
endothelia
may
serve
an
important
role
in
HIV-
1
tissue
invasion
and
angiogenesis.
Materials
and
Methods:
The
expression
of
chemokine
receptors
in
human
brain
microvascular
endothelial
cells
(BMVEC)
and
coronary
artery
endothelial
cells
(CAEC)
in
vitro
and
cryostat
sections
of
the
heart
tissue
was
determined
by
light
and
confocal
microscopy
and
flow
cytometry
with
monodonal
antibodies.
Chemotaxis
of
endothelia
by
CC
chemokines
was
evaluated
in
a
trans-
migration
assay.
Results:
In
BMVEC,
the
chemokine
receptors
CCR3
and
CXCR4
showed
the
strongest
expression.
CXCR4
was
localized
by
confocal
microscopy
to
both
the
cytoplasm
and
the
plasma
membrane
of
BMVEC.
In
CAEC,
CXCR4
demonstrated
a
strong
expression
with
predominantly
periplasmic
localization.
CCR5
expression
was
detected
both
in
BMVEC
and
CAEC
but
at
a
lower
level.
Human
umbilical
cord
endothelial
cells
(HUVEC)
expressed
strongly
CXCR4
but
only
weakly
CCR3
and
CCR5.
Two
additional
CC
chemokines,
CCR2A
and
CCR4,
were
de-
tected
in
BMVEC
and
CAEC
by
immunostaining.
Immu-
nocytochemistry
of
the
heart
tissues
with
monodonal
antibodies
revealed
a
high
expression
of
CXCR4
and
CCR2A
and
a
low
expression
of
CCR3
and
CCR5
on
coronary
vessel
endothelia.
Coronary
endothelia
showed
in
vitro
a
strong
chemotactic
response
to
the
CC
chemokines
RANTES,
MIP-la,
and
MIP-1,8.
Conclusions:
The
endothelia
isolated
from
the
brain
display
strongly
both
the
CCR3
and
CXCR4
HIV-
1
core-
ceptors,
whereas
the
coronary
endothelia
express
strongly
only
the
CXCR4
coreceptor.
CCR5
is
expressed
at
a
lower
level
in
both
endothelia.
The
differential
dis-
play
of
CCR3
on
the
brain
and
coronary
endothelia
could
be
significant
with
respect
to
the
differential
sus-
ceptibility
of
the
heart
and
the
brain
to
HIV-1
invasion.
In
addition,
CCR2A
is
strongly
expressed
in
the
heart
endothelium.
All
of
the
above
chemokine
receptors
could
play
a
role
in
endothelial
migration
and
repair.
796
Molecular
Medicine,
Volume
5,
Number
12,
December
1999
Introduction
Chemokines
(up
to
50)
are
members
of
the
che-
mokine
superfamily
that
were
initially
identified
as
chemotactic,
low-molecular-weight
peptides.
Chemokines
are
classified
into
CXC
(or
a),
CC
(or
(),
C,
and
CXXXC
families
(1).
Although
CXC
che-
mokines
with
the
ELR
motif
serve
as
chemoattrac-
tants
for
neutrophils
and
CC
chemokines
attract
primarily
monocytes
and
T
cells,
chemokine
dis-
tinction
based
on
leukocyte
specificity
has
become
blurred
(2).
Chemokine
receptors
belong
to
the
large
family
of
G
protein-coupled,
seven-trans-
membrane
domain
(also
called
serpentine)
recep-
tors
(3),
which
are
displayed
on
many
different
cell
types,
including
endothelia
(2).
Chemokines
or-
chestrate
monocyte
and
lymphocyte
migration
across
the
endothelial
lining
of
blood
vessels,
a
process
that
plays
a
central
role
in
pathological
states
such
as
atherosclerosis
(4),
Alzheimer's
dis-
ease
(5),
multiple
sclerosis
(6),
and
HIV-
1
enceph-
alitis
(7).
For
example,
monocyte
chemotactic
protein-
I
(MCP-
1)
is
a
pivotal
chemokine
for
monocyte
migration
in
atherosclerosis
(8),
whereas
in
multiple
sclerosis
the
chemokines
in-
terferon-y
(IFN-,y)
-inducible
protein
(IP-
10),
monokine
induced
by
IFN-y
(MIG),
and
regulated
on
activation,
normal
T
cell
expressed
and
secreted
(RANTES)
are
considered
critical
for
mononuclear
infiltration
(9).
Different
chemokines
induce
the
accumulation
of
macrophages
in
fatty
streaks
of
atherosclerosis
(10),
migration
of
monocytes
across
coronary
endothelia
in
coronary
atherosclerosis
(11,12),
and
monocyte
infiltration
of
the
brain
in
AIDS
encephalitis
(13).
In
contrast
to
the
detailed
knowledge
of
the
multiple
effects
of
chemokines
on
leukocyte
mi-
gration
and
signaling
through
chemokine
recep-
tors
(14),
the
role
of
chemokine
receptors
on
endothelia
is
just
beginning
to
be
unraveled.
The
existence
of
chemokine
receptors
on
endothelia
has
been
obscured
by
the
presence
of
chemokine
binding
sites
that
do
not
display
the
chemokine
subclass
specificities
(
15)
and
may
represent
pro-
teoglycans
(16,17).
Specific
chemokine
binding
sites
on
human
brain
microvessels
were
visual-
ized
using
a
biotinylated
chemokine
binding
as-
say
(18).
Recently,
the
presence
of
CXCR4
on
human
umbilical
cord
endothelial
cells
(HUVEC)
was
shown
using
Northern
blotting,
immunocy-
tochemistry,
and
in
situ
hybridization
(19-21).
Some
of
these
studies
also
suggest
endothelial
expression
of
other
chemokine
receptors,
such
as
CCR3,
CCR1,
CCR2,
and
CXCR1
(20),
and
CCR1,
CCR2,
and
CCR5
(18).
Chemokines
and
chemokine
receptors
play
an
important
role
in
normal
development
and
inflammatory
and
angiogenic
responses
of
endo-
thelia.
In
the
development
of
the
vascular
sys-
tem,
CXCR4,
with
its
cognate
ligand
stromal
cell-
derived
factor-
I
(SDF-
1),
is
essential
for
vas-
cularization
of
the
gastrointestinal
tract
(22).
CXCR4
is
also
involved
in
B
cell
lymphopoiesis,
bone
marrow
myelopoiesis,
and
cardiac
ventric-
ular
septum
formation
(23).
CXC
chemokines
with
the
ELR
motif,
but
not
RANTES
or
MCP-
1,
have
a
potent
chemotactic
activity
on
bovine
adrenal
gland
endothelia
in
vitro
and
in
vivo,
although
the
binding
sites
on
endothelia
have
not
been
determined
(24).
Endothelial
CXCR4
responds
to
its
ligand,
SDF-
1,
by
mobilization
of
intracellular
calcium
and
endothelial
cell
migra-
tion
(20,19).
Endothelial
cells
from
different
sites
show
significant
heterogeneity
in
antigenic
properties
(25),
function
(26),
and
morphology
(27).
The
data
presented
by
Gupta
et
al.
(20)
and
this
com-
munication
suggest
that
endothelia
may
also
dif-
fer
in
chemokine
receptor
display.
Since
the
dis-
covery
in
HUVEC
of
a
predominant
message
for
CXCR4,
there
has
been
speculation
about
the
role of
endothelial
chemokine
receptors
and
their
modulation
by
viral
proteins
and
cytokines
in
HIV-1
invasion
into
tissues
(20,28).
CXCR4
transcription
in
endothelia
is
modulated
by
sev-
eral
cytokines,
including
IFN-,y,
interleukin-
1
3
(IL-1X3),
and
tumor
necrosis
factor-a
(TNF-a)
which
are
elevated
in
AIDS
(20,21).
Further-
more,
the
viral
protein
Tat
induces
dramatic
up-
regulation
of
CXCR4
and
CCR5
in
monocyte/
macrophages,
which
transmigrate
across
the
blood-brain
barrier
in
HIV-1
encephalitis
(29).
The
expression
of
the
HIV-
1
chemokine
corecep-
tors
on
endothelia
could
be
crucial
for
viral
in-
vasion
into
the
brain
that
may
occur
either
through
paracellular
viral
transport
(30,31)
or
by
a
transcellular
route
(C.
Gujuluva
et
al.,
unpub-
lished
results).
Furthermore,
we
speculate
that
the
receptors
could
be
involved
in
the
regulation
of
monocyte/macrophage
transmigration
across
the
blood-brain
barrier
and
the
blood-brain
bar-
rier
permeability.
In
this
work,
we
have
investigated
the
pres-
ence
of
CC
and
CXC
chemokine
receptors
in
cultured
endothelia
derived
from
the
coronary
Address
correspondence
and
reprint
requests
to:
Dr.
Milan
Fiala,
Department
of
Neurology,
UCLA
School
of
Medicine,
710
Westwood
Plaza,
Los
Angeles,
CA
90095-1769,
U.S.A.
Phone:
310-206-6392;
Fax:
310-794-5094;
E-mail:
fiala@ucla.edu
0.
Berger
et
al.:
Chemokine
Receptors
on
Endothelia
797
vessels
and
brain
microvessels,
and
in
coronary
endothelia
in
the
heart
tissues.
Both
coronary
artery
endothelial
cells
(CAEC)
and
brain
micro-
vascular
endothelial
cells
(BMVEC)
display
strongly
CXCR4,
as
described
by
others,
but
BM-
VEC
differ
from
CAEC
in
a
strong
expression
of
CCR3.
CCR5
is
expressed
at
a
lower
level
in
both
endothelia.
As
shown
by
confocal
microscopy,
CCR5
and
CXCR4
are
localized
both
on
the
plasma
membrane
and
in
the
cytoplasm
but
the
cytoplasmic
localization
is
stronger
in
BMVEC
than
in
CAEC.
CAEC
migrate
in
response
to
the
CC
chemokines
RANTES,
macrophage
inflam-
matory
protein-I
a
(MIP-
1
a),
and
macrophage
inflammatory
protein-
1/3
(MIP-
1,B).
Materials
and
Methods
Cells
CAEC
(Clonetics/BioWhittaker,
San
Diego,
CA)
were
isolated
by
the
producer
from normal
hu-
man
heart
tissues
by
dissection
of
coronary
ar-
teries
and
enzymatic
digestion
and
were
propa-
gated
in
75
cm2
T.C.
flasks
(Falcon
Plastics/
Becton
Dickinson
Labware,
Franklin
Lakes,
NJ)
using
EGMR-2-MV
Bullet
Kit
medium
(Clonet-
ics/BioWhittaker).
Adult
BMVEC
were
prepared
from
surgical
brain
tissues
obtained
during
removal
of
epilep-
togenic
foci
that
were
dissociated
and
cultured
by
the
techniques
described
previously
(32).
The
outgrowing
cells
were
purified
by
fluorescence-
activated
cell
sorting
(FACS)
using
uptake
of
acetylated
low-density
lipoprotein
labeled
with
1,1'
-dioctadecyl-
3,3,3',3
'tetramethylindocarbo-
cyanine
perchlorate
(DiI-Ac-LDL)
(Biomedical
Technologies,
Stoughton,
MA)
as
described
pre-
viously
(32).
BMVEC
were
propagated
in
75
cm2
T.C.
flasks
using
RPMI
1640
medium
(Irvine
Sci-
entific,
Santa
Ana,
CA)
with
10%
fetal
bovine
serum
(FBS),
10%
NuSerum
(Collaborative
Re-
search,
Bedford,
MA),
sodium
pyruvate,
and
modified
Eagle's
medium
(MEM)
nonessential
amino
acids
and
vitamins.
Three
different
strains
(two
from
adult
brain
and
one
from
a
pediatric
brain)
were
examined
for
chemokine
receptors.
HUVEC
were
prepared
by
harvesting
umbil-
ical
veins
by
collagenase
perfusion
(33),
and
were
pooled
and
plated
in
75
cm2
T.C.
flasks
pretreated
with
0.2%
gelatin
(Difco,
Detroit,
MI).
HUVEC
were
cultured
in
a
1:1
mixture
of
M199
(Gibco
BRL)
and
astrocyte-conditioned
medium
was
supplemented
with
10%
FBS
and
10%
bovine
calf
serum
(BCS)
(Hyclone
Labora-
tories),
1%
penicillin-streptomycin
(Gibco
BRL),
1
%
fungizone
(Gibco
BRL),
1
%
HEPES
buffer
(Gibco
BRL),
1
mg/ml
heparin
(Sigma),
and
50
mg/ml
endothelial
cell
growth
supplement
(Col-
laborative
Biomedical
Products,
Bedford,
MA)
(34).
The
endothelia
were
removed
from
flasks
using
0.05%
trypsin/5
mM
EDTA
(Gibco
BRL),
seeded
onto
glutaraldehyde-cross-linked,
gela-
tin-coated
coverslips
(34)
in
24-well
plates,
and
were
grown
to
confluence
in
the
medium
appro-
priate
for
each
cell
strain.
CAEC,
BMVEC,
and
HUVEC
were
stained
100%
positively
with
the
antibody
to
von
Willebrand
factor
and
vimentin
and
were
unstained
with
antibody
to
a-smooth
muscle
actin
(all
from
DAKO,
Carpinteria,
CA).
Antibodies
Monoclonal
antibody
(MAb)
anti-CXCR4
(clones
44708.111,
44716.111,
44717.111)
(R&D
Sys-
tems),
MAb
anti-CCR3
(7B
11)
(LeukoSite),
MAb
anti-CCR5
(clones,
45502.111,
45523.111,
45549.111,
and
45531.111)
(R&D
Systems),
MAb
anti-CXCRI
(also
designated
as
IL-8RA),
and
MAb
anti-CXCR2
(also
designated
IL-8RB)
were
from
Pharmingen
International
(La
Jolla,
CA).
Goat
polyclonal
antibodies
(PAb),
anti-
CCR1,
anti-CCR2A,
anti-CCR2B,
anti-CCR3,
an-
ti-CCR4,
anti-CCR5,
anti-CXCR3,
and
anti-
CXCR-4
(also
designated
anti-fusin)
were
from
Santa
Cruz
Biotechnology
(Santa
Cruz,
CA).
The
Santa
Cruz
goat
antibodies
were
prepared
with
the
following
peptides:
LERVSSTSPSTGEHELSA-
GF
(CCR1),
KSIGRAPEASLQDKEGA
(CCR2A),
DGVTSTNTPSTGEQEVSAGL
(CCR2B),
LERTSS-
VSPSTAEPELSIVF
(CCR3),
DTPSSSYTQSTMDH-
DLHDAL
(CCR4),
ERASSVYTRSTGEQEISVGL
(CCR5),
SSSRRDSSWSETSEASYSGL
(CXCR3),
and
ALTSVSRGSSLKILSKG
(CXCR4).
CCR2A
staining
of
the
coronary
vessels
was
inhibited
with
the
peptide
KSIGRAPEASLQDKEGA.
The
antibodies
produced
by
R&D
Systems
and
LeukoSite
were
obtained
through
the
Na-
tional
Institutes
of
Health
(NIH)
AIDS
Research
and
Reference
Reagent
Program.
Immunofluorescence
and
Immunocytochemistry
The
30,000
cells
were
grown
to
confluence
for
4
days
on
gelatin-coated,
glutaraldehyde
cross-
linked
coverslips
(34).
After
fixation
for
30
min
with
3
%
paraformaldehyde
at
40C,
the
cells
were
permeabilized
with
0.1%
Triton,
incubated
for
1
hr
with
a
primary
antibody
(MAb
or
PAb
at
5
798
Molecular
Medicine,
Volume
5,
Number
12,
December
1999
,ug/ml),
washed,
and
then
incubated
with
FITC-
conjugated
secondary
antibody.
For
dual
stain-
ing,
two
primary
mouse
and
rabbit
antibodies
and
two
secondary
FITC-
or
Texas
Red-conju-
gated
antibodies
specific
for
mouse
or
goat
im-
munoglobulin
G
(IgG)
were
used.
Immunofluo-
rescent
preparations
were
viewed
using
either
an
Olympus
Research
microscope
with
a
mercury
lamp
or a
Zeiss
LSM
310
confocal
laser
micro-
scope.
Immunocytochemical
staining
of
the
en-
dothelia
was
done
using
the
DAKO
LSAB+
Per-
oxidase
kit
(DAKO)
as
described
previously
(35).
Flow-Cytometric
Analysis
of
Chemokine
Receptors
The
endothelial
cells
were
removed
from
the
flask
using
the
Cell
Dissociation
Buffer
Enzyme-
Free
(Life
Technologies,
Gaithersburg,
MD).
All
staining
steps
were
performed
on
ice.
The
endo-
thelial
cells
were
washed
with
phosphate-buff-
ered
saline
(PBS)/1%
bovine
serum
albumin
(BSA)
by
centrifugation.
The
cells
were
then
re-
suspended
in
PBS/1%
BSA
and
stained
with
2
,ug/ml
of
each
primary
antibody
(CXCR4
MAb,
CCR5
MAb,
CXCR4
MAb,
or
IgG
isotype
control)
for
30
min.
After
two
washes
with
PBS/1
%
BSA,
rabbit
anti-mouse
FITC-antibody
was
added
for
30
min.
The
cells
were
washed
two
times
and
resuspended
in
PBS/1%
BSA.
Finally,
the
cells
were
fixed
with
2%
paraformaldehyde.
Receptor
expression
on
the
endothelia
was
analyzed
by
EPICS
XL-MCO
(Coulter).
Histochemistry
Fresh
myocardium
from
an
explanted
heart
of
two
patients
with
dilated
cardiomyopathy
was
obtained
from
the
Surgical
Pathology
Division
of
the
UCLA
Department
of
Pathology.
Specimens
were
embedded
in
O.C.T.
embedding
medium
from
Sakura
Finetek
U.S.A.
(Torrance
CA)
and
snap
frozen
in
liquid
nitrogen
and
dry
ice.
Cry-
ostat
sections
were
fixed
in
4%
paraformalde-
hyde
in
0.1
M
PBS
for
30
min
at
40C.
After
washing
in
PBS
the
slides
were
incubated
with
blocking
buffer
containing
0.5%
BSA
and
then
with
the
primary
antibody
diluted
1:200
in
PBS
with
0.1%
Triton
X-100
(Sigma)
for
12-18
hr
at
4°C.
As
the
primary
antibody,
mouse
MAb
to
CCR3,
CXCR4,
or
CCR5
(R&D),
or
goat
PAb
to
CCR1,
CCR2A,
CCR2B,
CCR3,
CCR4,
or
CCR5
(Santa
Cruz
Biotechnology)
was
used.
The
slides
were
washed
in
PBS
containing
Tween
20
(PB
ST)
(Sigma)
for
5
min
and
incubated
sequen-
tially
with
biotinylated
rabbit
anti-mouse
and
biotinylated
swine
anti-rabbit
antisera
(both
from
DAKO)
for
30
min
at
room
temperature.
Slides
were
then
washed
in
PBST
and
incubated
with
streptavidin-horseradish
peroxidase
(DAKO)
for
30
min.
Following
the
diaminobenzidine
reaction,
the
slides
were
counterstained
with
hematoxylin.
In
the
case
of
the
goat
PAb,
the
second
step
con-
sisted
of
biotin-conjugated
anti-goat
antibody.
This
was
followed
by
avidin
and
biotinylated
horserad-
ish
peroxidase
(ABC
Staining
System,
Santa
Cruz
Biotechnology).
Endothelial
Chemotaxis
Assay
Chemotaxis
of
endothelia
by
CC
chemokines
was
tested
in
96-well
chemotaxis
chambers
(NeuroProbe,
Gaithersburg,
MD),
and
50
,ul
of
EGMR-2-MV
Bullet
Kit
medium
(Clonetics/Bio-
Whittaker)
with
RANTES,
MIP-l1a,
or
MIP-1,B
at
the
indicated
concentration
was
placed
in
the
lower
chamber
and
1000
endothelial
cells
in
20
,ul
medium
was
introduced
in
the
upper
well
above
the
plate
(with
8-,um
pores)
separating
the
chambers.
The
chamber
was
incubated
at
370C
for
48
hr,
then
the
plate
was
removed
and
the
cells
on
the
upper
surface
were
removed
using
a
scraper.
The
lower
surface
of
the
plate
was
stained
using
0.1%
crystal
violet
and
the
cells
were
enumerated
using
10OX
magnification
in
a
horizontal
strip
across
each
well.
The
number
of
cells
was
multiplied
by
4
to
obtain
the
total
num-
ber
of
transmigrated
cells.
Results
Coronary
and
Brain
Endothelia
Display
CCR3,
CXCR4,
and
CCR5
In
Vitro
We
investigated
the
expression
of
chemokine
re-
ceptors
in
cultured
endothelia
using
immunofluo-
rescence
and
immunoperoxidase
techniques
with
monodonal
antibody
(MAb)
(CXCR1,
CXCR2,
CXCR4,
CCR3,
and
CCR5)
and/or
polydonal
anti-
body
(PAb)
(CXCR3,
CXCR4,
CCR1,
CCR2A,
CCR2B,
CCR3,
CCR4,
and
CCR5).
The
specificity
of
each
reaction
was
shown
by
the
absence
of
staining
when
the
primary
antibody
to
a
chemo-
kine
receptor
was
omitted
or
substituted
with
an
isotype-matched
IgG
(Fig.
1J-L).
CAEC
and
BM-
VEC
showed
strongly
positive
staining
by
the
im-
munofluorescence
(Fig.
IA-I)
and
the
immuno-
peroxidase
technique
(not
shown)
with
anti-
CCR3,
-CXCR4,
and
-CCR5.
The
staining
of
BMVEC
and
CAEC
was
consistently
greater
when
the
cells
were
fixed
before
the
staining
than
when
0.
Berger
et
al.:
Chemokine
Receptors
on
Endothelia
I
Fig.
1.
Immunofluorescent
stainiing
of
CCR3,
CCR5,
and
CXCR4
chemokine
receptors
in
BMVEC
and
CAEC.
Human
brain
(A,
D,
G,
J)
and
coronary
(B,
C,
E,
F,
H,
I,
K,
L)
endothelia
were
grown
on
coverslips
coated
with
glutaraldehyde-
fixed
gelatin
(34).
Confluent
monolayers
were
stained
by
the
immunofluorescent
technique
using
MAb
against
CCR3
(IgG2A
isotype)
(A-C),
CCR5
(IgG2B
isotype)
(D-F),
CXCR4
(IgG2A
isotype)
(G-
I),
or
an
IgG
mouse
isotype
antibody,
IgG2A
(J,
K),
or
IgG2B
(L).
The
cells
were
either
permeabilized
first
with
Triton
(A,
B,
D,
E,
G,
H,
J,
K),
or
they
were
stained
without
permeabilization
(C,
F,
I,
1).
The
IgG
isotype-stained
samples
were
photographed
using
a
six
times
longer
exposure
(compared
to
MAb-stained
preparations)
to
demonstrate
the
back-
ground
autofluorescence
staining
of
the
endothelia.
they
were
stained
without
fixation
(compare
Fig.
lB
with
Fig.
IC,
Fig.
IE
with
Fig.
IF,
and
Fig.
IH
with
Fig.
11).
In
addition,
two
CC
chemo-
kine
receptors
(CCR2A
and
CCR4)
were
positively
stained
by
inmmunocytochemistry
in
both
CAEC
and
BMVEC,
and
two
CXC
chemokine
receptors
(CXCR1
and
CXCR3)
were
stained
in
BMVEC
but
not
in
CAEC
(data
not
shown).
CXCR2
was
nega-
tive
by
immunocytochemical
staining
in
CAEC
and
BMVEC
(data
not
shown).
Laser
confocal
microscopic
examination
of
CAEC
revealed
CXCR4
(Fig.
2A)
and
CCR5
(Fig.
2B)
(green)
in
a
patchy
distribution
along
the
plasma
membrane
and
von
Willebrand
factor
(VWF)
(red)
in
a
granular-to-homogeneous
pat-
tern
in
the
perinuclear
location.
In
contrast
to
this
pattern
seen
in
CAEC,
in
BMVEC
both
CCR5
and
CXCR4
were
expressed
in
the
cytoplasm
and
on
the
plasma
membrane.
CXCR4
(Fig.
3B)
was
strongly
expressed
and
showed
substantial
colo-
calization
with
VWF
in
the
cytoplasm.
CCR5
ex-
pression
was
weak
(Fig.
3A).
799
800
Molecular
Medicine,
Volume
5,
Number
12,
December
1999
Fig.
3.
Confocal
microscopy
of
the
subcellular
localization
of
CCR5
(A)
and
CXCR4
(B)
in
BM-
VEC.
BMVEC
were
grown
on
coverslips
and
stained
as
described
in
Figure
2.
Fig.
2.
Confocal
microscopy
of
the
subcellular
localization
of
CXCR4
(A)
and
CCR5
(B)
in
CAEC.
CAEC
were
grown
on
coverslips
coated
with
glutaraldehyde-fixed
gelatin
(34).
The
cells
were
double
stained
using,
as
the
primary
antibody,
MAb
to
CXCR4
or
CCR5
followed
by
the
rabbit
antibody
to
von
Willebrand
factor
and,
as
the
secondary
anti-
body,
mouse
or
rabbit
IgG
labeled
with
FITC
or
Texas
Red,
respectively.
Chemokine
receptors
CCR5
and
CXCR4
appear
as
green
and
von
Willebrand
fac-
tor
as
red.
CCR3,
CXCR4,
and
CCR5
Receptor
Density
in
CAEC,
BMVEC,
and
HUVEC
The
expression
of
chemokine
receptors
in
BMVEC,
CAEC,
and
HUVEC
was
quantified
us-
ing
flow
cytometry
performed
with
the
cells
stained
without
prior
fixation.
In
the
case
of
BMVEC,
the
receptor
density
(%
staining
by
the
specific
antibody
-
%
staining
by
the
IgG
isotype
control)
was
determined
in
two
experiments
as
follows:
CCR3,
45%
and
77%;
CXCR4,
41%
and
51%;
and
CCR5,
4%
and
5%.
In
one
additional
experiment,
CXCR4
was
40%
and
CCR5
was
27%.
In
CAEC,
the
density
was
measured
in
two
experiments
with
the
following
results:
CCR3,
11°%
and
21%;
CXCR4,
41%
and
45%,
and
CCR5,
6%
and
4%.
In
HUVEC,
the
results
were
7
%
with
CCR3,
48%
with
CXCR4,
and
8%
with
CCR5.
The
flow-cytometric
diagrams
of
a
typical
experiment
illustrate
the
above
receptor
densi-
ties,
as
follows:
in
BMVEC
the
order
was
CCR3
=
CXCR4
>
CCR5
(Fig.
4A);
in
CAEC
the
order
was
CXCR4
>
CCR3
>
CCR5
(Fig.
4B);
and
in
HUVEC
the
order
was
CXCR4>
CCR5
and
CCR3
0.
Berger
et
al.:
Chemokine
Receptors
on
Endothelia
(A)
(B)
(C)
Blak
solid:
IpG
control
Black
open:
CCR3
Gray
solid:
CCRS
Gray
open:
CXCR44
_
A-
f__j
v
J
Lb-
w
*
*-w_
6
s
P~~~~~~~~~~~~~~~..
..VW~
Fig.
4.
Chemokine
receptor
expression
in
CAEC,
BMVEC,
and
HUVEC
evaluated
by
flow
cytometry.
Endothelial
cells
BMVEC
(A),
CAEC
(B),
and
HUVEC
(C)
were
lifted
from
the
flask
in
a
cell-dissociation
buffer,
washed,
and
stained
using
the
monoclonal
antibody
to
CCR3,
CCR5,
or
CXCR4,
and
analyzed
by
flow
cytometry
as
described
in
Ma-
terials
and
Methods.
(Fig.
4C).
When
the
cells
were
fixed
first
and
stained
second,
the
percentage
of
staining
in-
creased.
CCR3,
CXCR4,
CCR5,
and
CCR2A
Are
Expressed
in
Coronary
Endothelia
of
the
Heart
Immunohistochemical
staining
of
the
human
heart
sections
was
performed
with
three
heart
specimens.
Typical
results
revealed
a
strong
(4+)
staining
of
small
venules
and
capillaries
in
the
myocardium
with
goat
polyclonal
antiserum
to
CCR2A
(Fig.
SD),
a
moderately
strong
(3+)
staining
with
a
monoclonal
antibody
to
CXCR4
(Fig.
5C),
a
moderate
(2+)
staining
with
mono-
clonal
anti-CCR5
(Fig.
SB),
and
a
weak
(1+)
staining
with
mouse
MAb
to
CCR3
(Fig.
5A).
A
positive
staining
reaction
appeared
to
involve
the
endothelial
cell
cytoplasm
as
well
as
the
cell
membrane
(Fig.
SC,
D).
IgG
isotype
control
was
negative
(Fig.
SF).
Preincubation
of
anti-CCR2A
with
its
blocking
peptide
completely
inhibited
the
staining
reaction
(compare
Fig.
SD
with
Fig.
SE).
The
results
of
immunostaining
of
the
heart
tissues
with
anti-CCRI,
CCR2B,
and
CCR4
were
negative.
RANTES,
MIP-lIa,
and
MIP-1f3
Stimulate
Migration
of
Coronary
Endothelia
CAEC
were
tested
for
the
chemotactic
responses
to
the
CC
chemokines
RANTES,
MIP-la,
and
MIP-
I,B.
Each
of
these
chemokines
produced
en-
dothelial
cell
migration
at
a
concentration
above
12.5
,ug/ml
(Fig.
6).
A
second
experiment
con-
firmed
the
chemotactic
responses
to
these
che-
mokines
but
the
threshold
for
chemotactic
activ-
ity
was
one
dilution
lower
(12.5
,ug/ml)
with
each
chemokine.
Discussion
We
have
shown
that
human
coronary
and
brain
endothelia
express
to
a
different
degree
the
CC
chemokine
receptors
CCR3
and
CCR5,
and
the
CXC
receptor
CXCR4.
Whereas
in
CAEC
usually
only
CXCR4
achieved
a
high
density,
in
BMVEC,
CCR3
and
CXCR4
were
both
highly
expressed.
CCR5
was
expressed
at
a
low
density
in
both
endothelia.
The
effect
of
successive
in
vitro
pas-
sages
of
the
endothelia
appeared
to
decrease
the
chemokine
receptor
density.
The
pattern
of
a
strong
expression
of
CXCR4
and
a
weak
expres-
sion
of
CCR3
was
also
found
in
the
coronary
vessel
endothelia
in
the
heart.
Two
other
chemo-
kine
receptors,
CCR2A
and
CCR4,
were
detected
by
immunostaining
in
both
CAEC
and
BMVEC,
and
CXCR1
and
CXCR3
were
found
in
BMVEC
only.
In
addition,
CCRS
and
CXCR4
mRNAs
were
detected
in
BMVEC
by
reverse
transcriptase
polymerase
chain
reaction
(RT-PCR)
with
five
times
greater
expression
of
CXCR4
mRNA
(C.
Gujuluva
et
al.,
unpublished
results).
The
pres-
ence
of
multiple
chemokine
receptors
in
endo-
thelia
is
supported
by
the
RT-PCR
data
of
Gupta
et
al.
(20)
in
HUVEC
and
BMVEC,
which
showed
an
abundant
transcription
of
CXCR4
and
a
I
801
I
i
802
Molecular
Medicine,
Volume
5,
Number
12,
December
1999
Fig.
5.
Coronary
endothe-
lia
display
CCR2A,
CXCR4,
CCR5,
and
CCR3
(in
this
order).
Cryostat
sections
of
the
heart
were
stained
by
the
peroxidase
technique
(DAKO)
using
the
antibody
to
CCR3
(A),
CCR5
(B),
CXCR4
(C),
IgG
isotype
control
(F),
CCR2A
(D),
or
CCR2A
absorbed
with
the
peptide
KSIGRAPEASLQD-
KEGA
(E).
weaker
transcription
of
CXCR1,
CCR1,
and
CCR2.
A
recent
RT-PCR
study
of
HUVEC
noted
the
presence
of
CXC
chemokine
mRNAs
but
not
CC
chemokine
mRNAs
(21),
and
the
authors
speculated
that
endothelial
cells
do
not
express
CC
chemokine
receptors.
However,
their
conclu-
sions
were
based
on
transcriptional
studies
and
involved
only
HUVEC.
Our
flow-cytometric
data
demonstrate
that
whereas
CXCR4
and
CCR3
are
both
highly
expressed
in
BMVEC,
only
CXCR4
is
highly
expressed
in
CAEC
and
HUVEC.
These
results
were
subjected
to
quantitative
variation,
with
preservation
of
these
relationships
between
the
receptors,
due
to
the
effects
of
cell
passage
and
fixation.
The
fixation
before
staining
in-
creased
the
staining
density
because
of
staining
of
intracellular
receptors.
These
results
strongly
suggest
that
brain,
coronary,
and
umbilical
en-
dothelia
differ
from
each
other
with
respect
to
chemokine
receptor
expression.
The
histochem-
ical
study
of
the
heart
provided
further
evidence
of
the
relevance
of
chemokine
receptors
to
in-
flammatory
responses
of
coronary
endothelia,
as
four
chemokine
receptors,
CCR2A,
CCR3,
CCR5,
and
CXCR4,
were
found
on
the
coronary
vessels
but
only
CXCR4
and
CCR2A
were
expressed
at
a
high
level,
in
agreement
with
the
above
results.
Chemokine
HIV-
1
coreceptors,
CCR5
and
CXCR4,
play
a
crucial
role
in
HIV-
1
entry
into
the
blood
cells
(36).
On
leukocytes,
CXCR4
serves
as
a
coreceptor
for
T
cell
line
tropic
"X"
strains,
whereas
the
CC
chemokine
receptor
CCR5
is
a
main
coreceptor
for
M-tropic
"R"
strains.
In
blood-derived
dendritic
cells,
which
express
CXCR4
and
CCR5
mRNAs
(37),
the
en-
try
of
M-tropic
strains
is
inhibited
by
the
chemo-
kine
RANTES
and
the
entry
of
the
IIIB
strain
by
the
chemokine
SDF-1.
CD34+
progenitors
are
0.
Berger
et
al.:
Chemokine
Receptors
on
Endothelia
803
(A)
0
_
I
I
E
z
(B)
p
300
X
250
r
_
200
*a3150
3
100
E
z
50
(C)
5
I
E
z
0
12.5
25
50
100
RANTES
Concenftron
(nghnl)
0
12.5
25
50
MIP-l
a
Concentaton
(ngiml)
100
MIP-1p
Concenration
(nglml)
Fig.
6.
Endothelial
cell
migration
across
a
Neu-
roProbe
chamber
filter.
One
thousand
CAEC
were
seeded
on
top
of
each
well
in
a
NeuroProbe
chamber
(8
,um
pore
diameter)
with
each
tested
chemokine,
RANTES
(A),
MIP-la
(B),
or
MIP-113
(C),
at
the
indicated
concentration
in
the
lower
chamber.
The
endothelia
were
allowed
to
migrate
for
48
hr,
then
the
filter
was
removed
and
the
cells
on
the
top
of
each
well
were
scraped.
After
staining
by
crystal
violet,
the
transmigrated
cells
on
the
lower
surface
of
each
well
were
determined
by
a
mi-
croscopic
count
as
described
in
Materials
and
Meth-
ods.
susceptible
to
infection
by
various
HIV-1
strains
and
cognate
ligands
for
CXCR4
and
CCR5
mod-
ulate
HIV-1
infection
(38).
CC
and
CXC
chemokine
receptors
belong
to
a
large
family
of
cell
surface,
seven-transmem-
brane
domain,
G
protein-coupled
receptors
that
bind
and
signal
upon
binding
of
one
or
more
CC
or
CXC
chemokines
(3).
CCR2
is
a
receptor
pre-
viously
detected
in
monocytes,
dendritic
cells,
natural
killer
(NK)
cells,
and
T
lymphocytes,
but
not
in
neutrophils,
and
is
thus
considered
to
be
important
in
chronic
inflammation,
including
atherosclerosis.
CCR2
has
recently
been
shown
to
function
as
a
receptor
for
MCP-
1
on
endothe-
lia,
inducing
their
chemotactic
migration
and
possibly
endothelial
wound
repair
(39).
CCR3
is
found
in
brain
microglia,
in
which
it
mediates
HIV-1
entry
(40).
CCR5
is
a
major
HIV-1
core-
ceptor,
which
plays
a
crucial
role
in
HIV
infection
of
peripheral
blood-derived
dendritic
cells,
CD34+
hematopoeitic
progenitor
cells,
and
acti-
vated/memory
Thl
lymphocytes
(41,42).
CCR5
expression
on
freshly
isolated
T
cells
is
increased
by
activation
with
IL-2
and
other
stimuli
(41).
In
contrast
to
the
pivotal
significance
of
CCR5
on
blood
cells
for
HIV-1
infection
(as
well
as
other
infections),
the
role
of
CCR5
in
neurons,
astro-
cytes,
and
other
cells
has
not
been
established.
CXCR4
is
the
major
chemokine
coreceptor
for
T-tropic
HIV-1
strains
that
is
widely
distributed
on
most
hematopoietic
cell
types,
endothelial
cells
(20),
neurons
in
both
the
peripheral
and
central
nervous
systems
(43),
microglia,
and
as-
trocytes
(40).
CXCR4
appears
to
play
an
essential
role
in
development,
as
shown
in
mice
(22).
Our
studies
of
the
heart
tissues
showed
that
CCR2A
and
CXCR4,
and,
to
a
lesser
degree,
CCR3
and
CCR5
were
displayed
on
coronary
endothelia.
We
have
not
excluded
a
possibility
that
this
staining
pattern
is
related
to
the
health
status
of
each
donor,
as
the
hearts
were
removed
before
transplantation
of
patients
with
conges-
tive
cardiomyopathy.
The
receptors
were
distrib-
uted
diffusely
in
the
endothelial
cytoplasm
and
on
the
plasma
membrane,
in
agreement
with
the
confocal
microscopic
results
in
coronary
endo-
thelia.
The
receptors
were
not
limited
to
-the
abluminal
side
of
endothelia,
as
found
by
And-
jelkovic
et
al.
(18),
by
staining
isolated
microves-
sels
with
MAb
to
CCR1,
CCR3,
and
CCR5.
The
functional
role
of
chemokine
receptors
on
brain
endothelia,
in
particular
CCR3,
CXCR4,
and,
to
a
lesser
degree,
CCR5,
could
be
signifi-
cant
for
HIV-
1
neuroinvasion.
Macrophage-
tropic
strains
circulating
at
a
very
high
titer
(107
RNA
copies/ml
plasma)
during
the
primary
in-
fection
could
find
early
entry
into
the
brain
be-
cause
of
a
high
expression
on
BMVEC
of
CCR3
and
a
variable
CCR5
expression.
Our
recent
work
strongly
suggests
that
HIV
invades
across
brain
endothelia
via
both
the
paracellular
(30,31)
and
the
transcellular
route
(C.
Gujuluva
et
al.,
unpublished
results)
without
undergoing
replication
in
BMVEC.
However,
we
have
not
I
T
m
804
Molecular
Medicine,
Volume
5,
Number
12,
December
1999
yet
obtained
conclusive
evidence
for
the
role
of
chemokine
receptors
in
viral
transcytosis.
After
the
neuroinvasion,
the
macrophage-tropic
HIV-
1
strains
utilizing
CCR3
and
CCR5
could
effectively
enter
the
microglia
via
CCR3
(40)
and
CCR5
(44).
SIV
infects
simian
brain
endothelial
cells
by
a
CCR5-dependent
mechanism,
which
is
inhib-
ited
by
the
CCR5
ligand
RANTES
(45),
but
this
route
may
be
limited
in
human
brain
endothelia
because
of
the
lower
expression
of
CCR5.
The
presence
of
CXCR4
on
brain
endothelia
may
al-
low
the
T-tropic
strains
emerging
late
in
the
dis-
ease
to
infect
the
brain.
Coronary
endothelia,
unlike
brain
endothelia,
appear
to
have
a
re-
duced
expression
of
CCR3,
which
may
limit
the
ability
of
macrophage-tropic
strains
circulating
early
after
infection
to
infect
the
heart.
The
cor-
onary
endothelia,
however,
express
CXCR4
and
may
be
susceptible
to
T-tropic
strains
that
emerge
late
in
the
course
of
AIDS,
when
HIV-1
cardiomyopathy
is
apt
to
develop.
Acknowledgments
This
work
was
supported
in
part
by
NIH
grant
DA10442
and
a
subcontract
NS
26126
to
M.F.
and
M.C.G.,
HL
48493
to
M.H.W.,
and
RO-1
NS
26310
and
RO-1
HL
61951
to
K.-S.K.A.R.B.
is
supported
in
part
by
grants
from
NIH
HL-42550,
The
Methodist
Hospital
Foundation,
a
Chao
Fel-
lowship,
and
ALA
RG
068.
The
authors
acknowl-
edge
the
UCSD
Center
for
AIDS
Research
and
NIH
DAIDS
2
P30
AI36214-07.
We
thank
Philip
Murphy
for
incisive
comments,
Matthew
Schibler
for
assistance
with
confocal
microscopy,
and
Carol
Appleton
for
technical
assistance
with
illustrations.
References
1.
Luster
A.
(1998)
Chemokines-chemotactic
cyto-
kines
that
mediate
inflammation.
N.
Engl.
J.
Med.
338:
436-445.
2.
Taub
D.
(1996)
Chemokine-leukocyte
interac-
tions-the
voodoo
that
they
do
so
well.
Cytokine
Growth
Factors
Rev.
7:
355-376.
3.
Murphy
P.
(1996)
Chemokine
receptors:
struc-
ture,
function
and
role
in
microbial
pathogenesis.
Cytokine
Growth
Factor
Rev.
7:
47-64.
4.
Berliner
JA,
Territo
MC,
Sevanian
A,
et
al.
(1990)
Minimally
modified
low
density
lipoprotein
stim-
ulates
monocyte
endothelial
interactions.
J.
Clin.
Invest.
85:
1260-1266.
5.
Maat-Schieman
ML,
van
Duinen
SG,
Rozemuller
AJ,
Haan
J,
Roos
RA.
(1997)
Association
of
vas-
cular
amyloid
beta
and
cells
of
the
mononuclear
phagocyte
system
in
hereditary
cerebral
hemor-
rhage
with
amyloidosis
(Dutch)
and
Alzheimer
disease.
J.
Neuropathol.
Exp.
Neurol.
56:
273-284.
6.
Sorensen
TL,
Ransohoff
RM
(1998)
Etiology
and
pathogenesis
of
multiple
sclerosis.
Semin.
Neurol.
18:
287-294.
7.
Glass
JD,
Fedor
H,
Wesselingh
SL,
McArthur
JC.
(1995)
Immunocytochemical
quantitation
of
hu-
man
immunodeficiency
virus
in
the
brain:
corre-
lations
with
dementia.
Ann.
Neurol.
38:
755-762.
8.
Navab
M,
Imes
SS,
Hama
SY,
et
al.
(1991)
Mono-
cyte
transmigration
induced
by
modification
of
low
density
lipoprotein
in
cocultures
of
human
aortic
wall
cells
is
due
to
induction
of
monocyte
chemotactic
protein
1
synthesis
and
is
abolished
by
high
density
lipoprotein.
J.
Clin.
Invest.
88:
2039-2046.
9.
Sorensen
TL,
Tani
M,
Jensen
J,
et
al.
(1999)
Ex-
pression
of
specific
chemokines
and
chemokine
receptors
in
the
central
nervous
system
of
multi-
ple
sclerosis
patients.
J.
Clin.
Invest.
103:
807-815.
10.
Boisvert
WA,
Santiago
R,
Curtiss
LK,
Terkeltaub
RA.
(1998)
A
leukocyte
homologue
of
the
IL-8
receptor
CXCR-2
mediates
the
accumulation
of
macrophages
in
atherosclerotic
lesions
of
LDL
re-
ceptor-deficient
mice.
J.
Clin.
Invest.
101:
353-363.
11.
Ross
R.
(1999)
Atherosclerosis-an
inflammatory
disorder.
N.
Engl.
J.
Med.
340:
115-126.
12.
Terkeltaub
R,
Boisvert
WA,
Curtiss
LK.
(1998)
Chemokines
and
atherosclerosis.
Curr.
Opin.
Lipi-
dol.
9:
397-405.
13.
Schmidtmayerova
H,
Nottet
H,
Nuovo
G.
(1996)
HIV-
1
infection
alters
chemokine
beta-peptide
ex-
pression
in
human
monocytes:
implications
for
recruitment
of
leukocytes
into
brain
and
lymph
nodes.
Proc.
Soc.
Natl.
Acad.
Sci.
U.S.A.
93:
700-704.
14.
Richmond
A,
Mueller
S,
White
JR,
Schraw
W.
(1997)
C-X-C
chemokine
receptor
desensitization
mediated
through
ligand-enhanced
receptor
phosphorylation
on
serine
residues.
Methods
Enzy-
mol.
288:
3.
15.
Hub
E,
Rot
A.
(1998)
Binding
of
RANTES,
MCP-1,
MCP-3,
and
MIP-la
to
cells
in
human
skin.
Am.
J.
Pathol.
152:
749-757.
16.
Tanaka
Y,
Adams
DH,
Shaw
S.
(1993)
Proteogly-
cans
on
endothelial
cells
present
adhesion-induc-
ing
cytokines
to
leukocytes.
Immunol.
Today
14:
111-115.
17.
Luster
AD,
Greenberg
SM,
Leder
P.
(1995)
The
IP-10
chemokine
binds
to
a
specific
cell
surface
heparan
sulfate
site
shared
with
platelet
factor
4
and
inhibits
endothelial
cell
proliferation.
J.
Exp.
Med.
182:
219-231.
18.
Andjelkovic
AV,
Spencer
DD,
Pachter
JS.
(1999)
Visualization
of
chemokine
binding
sites
on
hu-
man
brain
microvessels.
J.
Cell
Biol.
145:
403-412.
19.
Volin
M,
Joseph
L,
Shockley
M,
Davies
P.
(1998)
Chemokine
receptor
CXCR4
expression
in
endo-
0.
Berger
et
al.:
Chemokine
Receptors
on
Endothelia
805
thelium.
Biochem.
Biophys.
Res.
Commun.
242:
46-
53.
20.
Gupta
S,
Lysko
P,
Pillarisetti
K,
Ohlstein
E,
Stadel
J.
(1998)
Chemokine
receptors
in
human
endo-
thelial
cells.
J.
Biol.
Chem.
273:
4282-4287.
21.
Feil
C,
Augustin
HG.
(1998)
Endothelial
cells
dif-
ferentially
express
functional
CXC-chemokine
re-
ceptor-4
(CXCR-4/fusin)
under
the
control
of
au-
tocrine
activity
and
exogenous
cytokines.
Biochem.
Biophys.
Res.
Commun.
247:
38-45.
22.
Tachibana
K,
Hirota
S,
Iizasa
H,
et
al.
(1998)
The
chemokine
receptor
CXCR4
is
essential
for
vascu-
larization
of
the
gastrointestinal
tract
[see
com-
ments].
Nature
393:
591-594.
23.
Nagasawa
T,
Hirota
S,
Tachibana
K,
et
al.
(1996)
Defects
of
B-cell
lymphopoiesis
and
bone-marrow
myelopoiesis
in
mice
lacking
the
CXC
chemokine
PBSF/SDF-1.
Nature
382:
635-638.
24.
Strieter
RM,
Polverini
PJ,
Kunkel
SL,
et
al.
(1995)
The
functional
role
of
the
ELR
motif
in
CXC
che-
mokine-mediated
angiogenesis.
J.
Biol.
Chem.
270:
27348-27357.
25.
Page
C,
Rose
M,
Yacoub
M,
Pigott
R.
(1992)
An-
tigenic
heterogeneity
of
vascular
endothelium.
Am.
J.
Pathol.
141:
763-683.
26.
Gerritsen
M.
(1987)
Functional
heterogeneity
of
vascular
endothelial
cells.
Biochem.
Pharmacol.
36:
2701-2711.
27.
Craig
L,
Spelman
J,
Strandberg
J,
Zink
M.
(1998)
Endothelial
cells
from
diverse
tissues
exhibit
dif-
ferences
in
growth
and
morphology.
Microvasc.
Res.
55:
65-76.
28.
Vicenzi
E,
Biswas
P,
Mengozzi
M,
Poli
G.
(1997)
Role
of
pro-inflammnatory
cytokines
and
beta-che-
mokines
in
controlling
HIV
replication.
J.
Leuko-
cyte
Biol.
62:
34-40.
29.
Huang
L,
Bosch
I,
Hofmann
W,
Sodroski
J,
Pardee
A.
(1998)
Tat
protein
induces
HIV-
1
coreceptors
and
promotes
infection
with both
macrophage-
tropic
and
T-lymphotropic
HIV-
1
strains.
J.
Virol.
72:
8952-8960.
30.
Fiala
M,
Looney
DJ,
Stins
M,
et
al.
(1997)
TNF-
alpha
opens
a
paracellular
route
for
HIV-1
inva-
sion
across
the
blood-brain
barrier.
Mol.
Med.
3:
553-564.
31.
Zhang
L,
Looney
D,
Taub
D,
et
al.
(1998)
Cocaine
opens
the
blood-brain
barrier
to
HIV-1
invasion.
J.
Neurovirol.
4:
619-626.
32.
Stins
M,
Gilles
F,
Kim
K.
(1997)
Selective
expres-
sion
of
adhesion
molecules
on
human
brain
mi-
crovascular
endothelial
cells.
J.
Neuroimmunol.
76:
81-90.
33.
Huang
AJ,
Furie
MB,
Nicholson
SC,
Fischbarg
J,
Liebovitch
LS,
Silverstein
SC.
(1988)
Effects
of
human
neutrophil
chemotaxis
across
human
en-
dothelial
cell
monolayers
on
the
permeability
of
these
monolayers
to
ions
and
macromolecules.
J.
Cell.
Physiol.
135:
355-366.
34.
Burns
AR,
Walker
DC,
Brown
ES.
(1997)
Neutro-
phil
transendothelial
migration
is
independent
of
tight
junctions
and
occurs
preferentially
at
tricel-
lular
corners.
J.
Immunol.
159:
2893-2903.
35.
Fiala
M,
Rhodes
RH,
Shapshak
P,
et
al.
(1996)
Regulation
of
HIV-1
infection
in
astrocytes:
ex-
pression
of
Nef,
TNF-a
and
IL-6
is
enhanced
in
coculture
of
astrocytes
with
macrophages.
J.
Neu-
rovirol.
2:158-166.
36.
Berger
E.
(1998)
Introduction:
HIV
co-receptors
solve
old
questions
and
raise
many
new
ones.
Immunology
10:
165-168.
37.
Granelli-Piperno
A,
Moser
B,
Pope
M,
et
al.
(1996)
Efficient
interaction
of
HIV-
1
with
purified
dendritic
cells
via
multiple
chemokine
receptors.
J.
Exp.
Med.
184:
2433-2438.
38.
Ruiz
M,
Cicala
C,
Arthos
J,
et
al.
(1998)
Peripheral
blood-deriverd
CD34+
progenitor
cells:
CXC
che-
mokine
receptor
4
and
CC
chemokine
receptor
5
expression
and
infection
by
HIV.
J.
Immunol.
161:
4169-4176.
39.
Weber
KS,
Nelson
PJ,
Grone
HJ,
Weber
C.
(1999)
Expression
of
CCR2
by
endothelial
cells:
implica-
tions
for
MCP-1
mediated
wound
injury
repair
and
In
vivo
inflammatory
activation
of
endothe-
lium.
Arterioscler.
Thromb.
Vasc.
Biol.
19:
2085-2093.
40.
He
J,
Chen
Y,
Farzan
M,
et
al.
(1997)
CCR3
and
CCR5
are
co-receptors
for
HIV-1
infection
of
mi-
croglia.
Nature
385:
645-649.
41.
Bleul
CC,
Wu
L,
Hoxie
JA,
Springer
TA,
Mackay
CR.
(1997)
The
HIV
coreceptors
CXCR4
and
CCR5
are
differentially
expressed
and
regulated
on
hu-
man
T
lymphocytes
[see
comments].
Proc.
Natl.
Acad.
Sci.
U.S.A.
94:
1925-1930.
42.
Loetscher
P,
Uguccioni
M,
Bordoli
L,
et
al.
(1998)
CCR5
is
characteristic
of
Thl
lymphocytes
[letter].
Nature
391:
344-345.
43.
Hesselgesser
J,
Halks-Miller
M,
DelVecchio
V,
et
al.
(1997)
CD4-independent
association
between
HIV-1
gp120
and
CXCR4:
functional
chemokine
receptors
are
expressed
in
human
neurons.
Curr.
Biol.
7:112-121.
44.
Albright
A,
Shieh
JT,
Itoh
T,
et
al.
(1999)
Micro-
glia
express
CCR5,
CXCR4,
and
CCR3,
but
of
these,
CCR5
is
the
principal
coreceptor
for
HIV-1
dementia
isolates.
J.
Virol.
73:
205-223.
45.
Edinger
AL,
Mankowski
JL,
Doranz
BJ,
et
al.
(1997)
CD4-independent,
CCR5-dependent
infec-
tion
of
brain
capillary
endothelial
cells
by
a
neu-
rovirulent
simian
immunodeficiency
virus
strain.
Proc.
Natl.
Acad.
Sci.
U.S.A.
94:
14742-14747.
... ECs secrete CCL5 and constitutively express CCL5 receptors (CCR3/CCR5) on their surfaces; however, the effects of CCL5 on hBMEC responses remain enigmatic (37). Secreted CCL5 reportedly forms a filamentous complex on the cell surface of ECs that directs the chemotaxis of immune cells to the endothelium (38). ...
... CCL5-CCR3/5 neutralization decreases ZIKV infection in hBMECs. The CCL5 receptors CCR3 and CCR5 are expressed on hBMECs, and ZIKV-infected hBMECs highly induce and secrete CCL5 (37). CCL5 addition to hBMECs prior to or simultaneously with ZIKV adsorption had no effect on the initial ZIKV infection of hBMECs (see Fig. S1 in the supplemental material). ...
... CCL5 is a well-studied chemokine that recruits, activates, and directs leukocyte survival responses through CCR3/CCR5 receptors (26,36,52). Remarkably, hBMECs also express CCR3 and CCR5, yet roles for CCL5 in regulating hBMEC responses remain largely unknown (37). Our findings suggest the potential for ZIKV-induced CCL5 to promote hBMEC survival and prompted studies defining the role of CCL5 in ZIKV persistence in hBMECs. ...
Article
Full-text available
Our findings demonstrate that CCL5 is required for ZIKV to persistently infect human brain ECs that normally protect neuronal compartments. We demonstrate that ZIKV-elicited CCL5 secretion directs autocrine hBMEC activation of ERK1/2 survival pathways via CCR3/CCR5, and inhibiting CCL5/CCR3/CCR5 responses prevented ZIKV persistence and spread.
... Previous studies have shown that human endothelial cells express CCR2, 38 and our findings regarding the antiangiogenic effects of a CCR2i on PDXs make sense. By contrast, our in vivo and in vitro experiments suggested that the CCR2i directly attenuated cancer cell viability. ...
Article
Full-text available
Background: Anti-angiogenic therapy with bevacizumab (BEV), an anti-VEGF antibody, plays a critical role in the treatment of ovarian cancer. However, despite an encouraging initial response, most tumors become resistant to BEV over time, and a new strategy that enables sustainable treatment using BEV is therefore needed. Methods: To overcome the resistance to BEV in patients with ovarian cancer, we performed a validation study of combination therapy with BEV (10 mg/kg) and the CCR2 inhibitor BMS CCR2 22 (20 mg/kg) (BEV/CCR2i) using 3 consecutive patient-derived xenografts (PDXs) of immunodeficient mice. Results: BEV/CCR2i demonstrated a significant effect of growth suppression in the BEV-resistant serous PDX and BEV-sensitive serous PDX compared with BEV (30.4% after the second cycle and 15.5% after the first cycle, respectively), and treatment cessation did not attenuate this effect. Tissue clearing and immunohistochemistry with an anti-α-SMA antibody suggested that BEV/CCR2i suppressed angiogenesis from the host mice more than BEV. In addition, human CD31 immunohistochemistry revealed that BEV/CCR2i decreased microvessels originating from the patients to a significantly greater degree than BEV. Regarding the BEV-resistant clear cell PDX, the effect of BEV/CCR2i was unclear during the first five cycles, but the following two cycles of increased-dose BEV/CCR2i (CCR2i 40 mg/kg) significantly suppressed tumor growth compared with BEV (28.3%) by inhibiting the CCR2B-MAPK pathway. Conclusions: BEV/CCR2i showed a sustained anticancer immunity-independent effect in human ovarian cancer that was more significant in serous carcinoma than in clear cell carcinoma.
Article
Full-text available
CCR5 is among the most studied chemokine receptors due to its profound significance in human health and disease. The notion that CCR5 is a functionally redundant receptor was challenged through the demonstration of its unique protective role in the context of West Nile virus in both mice and humans. In the nearly two decades since this initial discovery, numerous studies have investigated the role of CCR5 in the context of other medically important neurotropic flaviviruses, most of which appear to support a broad neuroprotective role for this receptor, although how CCR5 exerts its protective effect has been remarkably varied. In this review, we summarize the mechanisms by which CCR5 controls neurotropic flaviviruses, as well as results from human studies evaluating a genetic link to CCR5, and propose unexplored areas of research that are needed to unveil even more exciting roles for this important receptor.
Article
Full-text available
Diepoxybutane (DEB) is the most toxic metabolite of the environmental chemical 1,3‐butadiene. We previously demonstrated the occurrence of DEB‐induced p53‐mediated apoptosis in human lymphoblasts. The p53 protein functions as a master transcriptional regulator in orchestrating the genomic response to a variety of stress signals. Transcriptomic analysis indicated that C‐C chemokine ligand 4 (CCL4) gene expression was elevated in a p53‐dependent manner in DEB‐exposed p53‐proficient TK6 cells, but not in DEB‐exposed p53‐deficient NH32 cells. Thus, the objective of this study was to determine whether the CCL4 gene is a transcriptional target of p53 and deduce its role in DEB‐induced apoptosis in human lymphoblasts. Endogenous and exogenous wild‐type p53 transactivated the activity of the CCL4 promoter in DEB‐exposed lymphoblasts, but mutant p53 activity on this promoter was reduced by ∼80% under the same experimental conditions. Knockdown of the upregulated CCL4 mRNA levels in p53‐proficient TK6 cells inhibited DEB‐induced apoptosis by ∼45%–50%. Collectively, these observations demonstrate for the first time that the CCL4 gene is upregulated by wild‐type p53 at the transcriptional level, and this upregulation mediates apoptosis in DEB‐exposed human lymphoblasts.
Article
Full-text available
Stroke is the world’s second major cause of adult death and disability, resulting in the destruction of brain tissue and long-term neurological impairment; induction of neuronal plasticity can promote recovery after stroke. C–C chemokine receptor 5 (CCR5) can direct leukocyte migration and localization and is a co-receptor that can mediate human immunodeficiency virus (HIV) entry into cells. Its role in HIV infection and immune response has been extensively studied. Furthermore, CCR5 is widely expressed in the central nervous system (CNS), is engaged in various physiological activities such as brain development, neuronal differentiation, communication, survival, and learning and memory capabilities, and is also involved in the development of numerous neurological diseases. CCR5 is differentially upregulated in neurons after stroke, and the inhibition of CCR5 in specific regions of the brain promotes motor and cognitive recovery. The mechanism by which CCR5 acts as a therapeutic target to promote neurorehabilitation after stroke has rarely been systematically reported yet. Thus, this review aims to discuss the function of CCR5 in the CNS and the mechanism of its effect on post-stroke recovery by regulating neuroplasticity and the inflammatory response to provide an effective basis for clinical rehabilitation after stroke.
Article
CC chemokine receptor 3 (CCR3) belongs to the G protein-coupled receptor family and is highly expressed in eosinophils and basophils. CCR3 is essential for recruiting eosinophils into the lung. Moreover, CCR3 was found in the serum of colorectal cancer patients higher than in the control group. Therefore, CCR3 will be a useful target for asthma and colorectal cancer diagnosis. This study developed a specific and sensitive monoclonal antibody (mAb) for mouse CCR3 (mCCR3), which is useful for flow cytometry using the Cell-Based Immunization and Screening method. The established anti-mCCR3 mAb, C3Mab-3 (rat IgG2a, kappa), reacted with mCCR3-overexpressed Chinese hamster ovary-K1 (CHO/mCCR3) cells through flow cytometry. C3Mab-3 also reacted with P388 (mouse lymphoid neoplasma) and J774-1 (mouse macrophage-like) cells, which express mCCR3 endogenously. Kinetic analyses using flow cytometry indicated that KDs of C3Mab-3 for CHO/mCCR3, P388, and J774-1 cells were 4.3 × 10-8 M, 2.6 × 10-7 M, and 2.4 × 10-7 M, respectively. C3Mab-3 could be a valuable tool for elucidating mCCR3-related biological response using flow cytometry.
Article
One of the hallmarks of atherosclerosis is ongoing accumulation of macrophages in the artery intima beginning at disease onset. Monocyte recruitment contributes to increasing macrophage abundance at early stages of atherosclerosis. Although the chemokine CCL5 (RANTES) has been studied in atherosclerosis, its role in the recruitment of monocytes to early lesions has not been elucidated. We show that expression of Ccl5 mRNA, as well as other ligands of the CCR5 receptor (Ccl3 and Ccl4), is induced in the aortic intima of Ldlr−/− mice 3 weeks after the initiation of cholesterol-rich diet (CRD)-induced hypercholesterolemia. En face immunostaining revealed that CCL5 protein expression is also upregulated at 3 weeks of CRD. Blockade of CCR5 significantly reduced monocyte recruitment to 3-week lesions, suggesting that chemokine signaling through CCR5 is critical. However, we observed that Ccl5-deficiency had no effect on early lesion formation and CCL5-blockade did not affect monocyte recruitment in Ldlr−/− mice. Immunostaining of the lesions in Ldlr−/− mice and reciprocal bone marrow transplantation (BMT) of Ccl5+/+ and Ccl5−/− mice revealed that CCL5 is expressed by both myeloid and endothelial cells. BMT experiments were carried out to determine if CCL5 produced by distinct cells has functions that may be concealed in Ccl5−/−Ldlr−/− mice. We found that hematopoietic cell-derived CCL5 regulates monocyte recruitment and the abundance of intimal macrophages in 3-week lesions of Ldlr−/− mice but plays a minor role in 6-week lesions. Our findings suggest that there is a short window in early lesion formation during which myeloid cell-derived CCL5 has a critical role in monocyte recruitment and macrophage abundance.
Article
Full-text available
Although cyclooxygenase (COX) role in cancer angiogenesis has been studied, little is known about its role in brain angioplasticity. In the present study, we chronically infused mice with ketorolac, a non‐specific COX inhibitor that does not cross the blood–brain barrier (BBB), under normoxia or 50% isobaric hypoxia (10% O2 by volume). Ketorolac increased mortality rate under hypoxia in a dose‐dependent manner. Using in vivo multiphoton microscopy, we demonstrated that chronic COX inhibition completely attenuated brain angiogenic response to hypoxia. Alterations in a number of angiogenic factors that were reported to be COX‐dependent in other models were assayed at 24‐hr and 10‐day hypoxia. Intriguingly, hypoxia‐inducible factor 1 was unaffected under COX inhibition, and vascular endothelial growth factor receptor type 2 (VEGFR2) and C‐X‐C chemokine receptor type 4 (CXCR4) were significantly but slightly decreased. However, a number of mitogen‐activated protein kinases (MAPKs) were significantly reduced upon COX inhibition. We conclude that additional, angiogenic factor‐independent mechanism might contribute to COX role in brain angioplasticity, probably including mitogenic COX effect on endothelium. Our data indicate that COX activity is critical for systemic adaptation to chronic hypoxia, and BBB COX is essential for hypoxia‐induced brain angioplasticity. These data also indicate a potential risk for using COX inhibitors under hypoxia conditions in clinics. Further studies are required to elucidate a complete mechanism for brain long‐term angiogenesis regulation through COX activity. image
Article
Full-text available
Background Cardiovascular disease is currently the leading cause of death in patients with human immunodeficiency virus on combination antiretroviral therapy. Although the use of the protease inhibitor ritonavir has been associated with increased prevalence of cardiovascular disease, the underlying mechanisms remain ill‐defined. Herein, we tested the hypothesis that ritonavir‐mediated lipoatrophy causes endothelial dysfunction via reducing endothelial leptin signaling. Methods and Results Long‐term (4 weeks) but not short‐term (3 days) treatment with ritonavir reduced body weight, fat mass, and leptin levels and induced endothelial dysfunction in mice. Moreover, ritonavir increased vascular NADPH oxidase 1, aortic H 2 O 2 levels as well as interleukin‐1β, GATA3 (GATA binding protein 3), the macrophage marker (F4/80), and C‐C chemokine receptor type 5 (CCR5) expression. Reactive oxygen species scavenging with tempol restored endothelial function, and both NADPH oxidase 1 and CCR5 deletion in mice protected from ritonavir‐mediated endothelial dysfunction and vascular inflammation. Remarkably, leptin infusion markedly improved endothelial function and significantly reduced vascular NADPH oxidase 1, interleukin‐1β, GATA3, F4/80, and CCR5 levels in ritonavir‐treated animals. Selective deficiency in endothelial leptin receptor abolished the protective effects of leptin infusion on endothelial function. Conversely, selective increases in endothelial leptin signaling with protein tyrosine phosphatase deletion blunted ritonavir‐induced endothelial dysfunction. Conclusions All together, these data indicate that ritonavir‐associated endothelial dysfunction is a direct consequence of a reduction in adiposity and leptin secretion, which decreases endothelial leptin signaling and leads to a NADPH oxidase 1–induced, CCR5‐mediated reduction in NO bioavailability. These latter data also introduce leptin deficiency as an additional contributor to cardiovascular disease and leptin as a negative regulator of CCR5 expression, which may provide beneficial avenues for limiting human immunodeficiency virus infection.
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
Full-text available
Based on their ability to induce leukocyte chemotaxis and adhesion to endothelial cells (ECs), chemokines have been implicated in driving inflammatory leukocyte emigration. Recently, it was suggested that chemokines can accomplish their pro-emigratory role more effectively while being bound to the luminal surface of the ECs. Previously, such binding was demonstrated in situ in human skin for the prototype alpha-chemokine interleukin (IL)-8. Here we used an in situ binding assay to investigate the binding characteristics of several beta-chemokines in intact human skin. RANTES, MCP-1, and MCP-3 bound, similar to IL-8, in a specific saturable manner to the ECs of venules and small veins but not arteries or capillaries. RANTES inhibited MCP-1 and MCP-3 binding and vice versa, indicating that the EC binding sites are shared among these beta-chemokines; moreover, IL-8 and RANTES cross-competed for each other's binding, suggesting that the same chemokine binding sites are used by members of alpha- and beta-chemokine subfamilies. Conversely, MIP-1alpha did not bind to the ECs and did not compete for binding of RANTES. Analogous to IL-8, all of the tested beta-chemokines bound to the resident dermal cells. As a novel aspect of chemokine interaction with cells in normal skin, we observed specific, saturable binding of RANTES, MCP-1, and MCP-3 but not MIP-1alpha or IL-8 to the ECs of dermal afferent lymphatic vessels. RANTES, MCP-1, and MCP-3 also cross-competed for each other's binding to lymphatics, suggesting a common binding site with a novel chemokine binding profile. We suggest that the chemokine binding to the ECs of lymphatics may be involved in the process of leukocyte entry into the afferent lymphatic vessels.
Article
Full-text available
Incubation of cocultures of human aortic endothelial (HAEC) and smooth muscle cells (HASMC) with LDL in the presence of 5-10% human serum resulted in a 7.2-fold induction of mRNA for monocyte chemotactic protein 1 (MCP-1), a 2.5-fold increase in the levels of MCP-1 protein in the coculture supernatants, and a 7.1-fold increase in the transmigration of monocytes into the subendothelial space of the cocultures. Monocyte migration was inhibited by 91% by antibody to MCP-1. Media collected from the cocultures that had been incubated with LDL induced target endothelial cells (EC) to bind monocyte but not neutrophil-like cells. Media collected from cocultures that had been incubated with LDL-induced monocyte migration into the subendothelial space of other cocultures that had not been exposed to LDL. In contrast, media from separate cultures of EC or smooth muscle cells (SMC) containing equal number of EC or SMC compared to coculture and incubated with the same LDL did not induce monocyte migration when incubated with the target cocultures. High density lipoprotein HDL, when presented to cocultures together with LDL, reduced the increased monocyte transmigration by 91%. Virtually all of the HDL-mediated inhibition was accounted for by the HDL2 subfraction. HDL3 was essentially without effect. Apolipoprotein AI was also ineffective in preventing monocyte transmigration while phosphatidylcholine liposomes were as effective as HDL2 suggesting that lipid components of HDL2 may have been responsible for its action. Preincubating LDL with beta-carotene or with alpha-tocopherol did not reduce monocyte migration. However, pretreatment of LDL with probucol or pretreatment of the cocultures with probucol, beta-carotene, or alpha-tocopherol before the addition of LDL prevented the LDL-induced monocyte transmigration. Addition of HDL or probucol to LDL after the exposure to cocultures did not prevent the modified LDL from inducing monocyte transmigration in fresh cocultures. We conclude that cocultures of human aortic cells can modify LDL even in the presence of serum, resulting in the induction of MCP-1, and that HDL and antioxidants prevent the LDL induced monocyte transmigration.
Article
Leukocyte recruitment from the blood circulation into tissue is essential for effective immune responses, and is, consequently, carefully regulated. In this article Yoshiya Tanaka and co-workers describe a model in which proteoglycans on the luminal surface of endothelium capture pro-adhesive cytokines. These cytokines provide the adhesion-inducing signal to particular leukocyte subsets which initiates their transmigration.
Article
'Restricted' human immunodeficiency virus type (HIV-1) infection of astrocytes is recognized in vivo in some pediatric and adult AIDS brains and in vitro in a small proportion of transfected primary fetal astrocytes. We investigated the extent of HIV-1(JR-FL) expression in fetal astrocytes and macrophages cultivated alone or together. Peak HIV-1 p24 antigen titres in supernatant fluids of macrophage cultures were increased with monocyte/macrophages from certain donors and were higher when macrophages were cocultivated with astrocytes. Structural HIV-1 gene (gp 41 and pol) products (protein and mRNA) were observed only in macrophages. Ten days after HIV-1(JR-FL) infection, astrocytes in a monoculture were stained negative or only weakly positive (1-2+) for Nef, whereas in a coculture up to 100% of astrocytes displayed Nef staining (up to 4+) in the cytoplasm. The streptavidine-biotine-peroxidase technique with certain monoclonal antibodies to Nef (Ovod et al, 1992) was specific for infected astrocytes. The intensity of Nef staining was higher in astrocytes cultivated with monocyte/macrophages from certain donors. In the coculture, tumor necrosis factor-alpha (TNF-alpha) was expressed in the astrocyte cytoplasm earlier after coinfection with HIV-1 and cytomegalovirus (CMV) compared to infection with HIV-1 alone. Interleukin-6 (IL-6) was secreted spontaneously and transiently in uninfected cocultures, but in a prolonged fashion following HIV-1 and HIV-1/CMV infections. The interactions between HIV-1- and CMV-infected macrophages and astrocytes lead to upregulation of TNF-alpha and IL-6 and enhancement of productive HIV-1 infection of macrophages and of 'restricted' HIV-1 infection of astrocytes with implications for the pathogenesis of AIDS dementia.
Article
The advanced lesions of atherosclerosis represent the culmination of a specialized form of chronic inflammation followed by a fibroproliferative process that takes place within the intima of the affected artery. Proliferation of smooth muscle cells and generation of connective tissue occur. Proliferation results from interactions between arterial smooth muscle, monocyte-derived macrophages, T lymphocytes, and endothelium. The initial lesion of atherosclerosis, the fatty streak, begins as an accumulation of monocytederived macrophages and T lymphocytes, which adhere and migrate into the intima of the affected artery. Smooth muscle cells, which are present in the intima or which migrate into the intima from the media, then replicate. Monocyte-derived macrophages and T cells also replicate during lesion formation and progression due to the production of cytokines and growth-regulatory molecules. These molecules determine whether there is proliferation and lesion progression or inhibition of proliferation and lesion regression. Several growthregulatory molecules may play critical roles in this process, including platelet-derived growth factor (PGDF), transforming growth factor beta, fibroblast growth factor, heparinbinding epidermal growth factor-like growth factor, and others. PDGF may be one of the principal components in this process because protein containing the PDGF B-chain has been demonstrated within activated lesion macrophages during every phase of atherogenesis. The presence of this growth factor and its receptors on lesion smooth muscle cells creates opportunities for smooth muscle chemotaxis and replication. Smooth muscle proliferation depends upon a series of complex signals based upon cellular interactions in the local microenvironment of the artery. The intracellular signalling pathways for mitogenesis versus chemotaxis are being investigated for smooth muscle. The roles of the cytokines and growth-regulatory peptides involved in these cellular interactions represent critical points of departure for intervention and the development of new diagnostic methods. In addition, magnetic resonance imaging has been developed to demonstrate the fine structure of lesions of atherosclerosis in peripheral arteries not subject to cardiac motion. This noninvasive methodology holds great promise for the future of these approaches.
Article
THE chemokines are a large family of small, structurally related cytokines1,2. The physiological importance of most members of this family has yet to be elucidated, although some are inducible inflammatory mediators that determine leukocyte chemotaxis1–5. Pre-B-cell growth-stimulating factor/stromal cell-derived factor-1 (PBSF/SDF-1) is a member of the CXC group of chemokines6,7. PBSF/SDF-1 stimulates proliferation of B-cell progenitors in vitro 6 and is constitutively expressed in bone-marrow-derived stromal cells6,7. Here we investigate the physiological roles of PBSF/SDF-1 by generating mutant mice with a targeted disruption of the gene encoding PBSF/SDF-1. We found that mice lacking PBSF/SDF-1 died perinatally and that although the numbers of B-cell progenitors in mutant embryos were severely reduced in fetal liver and bone marrow, myeloid progenitors were reduced only in the bone marrow but not in the fetal liver, indicating that PBSF/SDF-1 is responsible for B-cell lymphopoiesis and bone-marrow myelopoiesis. In addition, the mutants had a cardiac ventricular septal defect. Hence, we have shown that the chemokine PBSF/SDF-1 has several essential functions in development.
Article
‘Restricted’ human immunodeficiency virus type (HTV-1) infection of astrocytes is recognized in vivo in some pediatric and adult AIDS brains and in vitro in a small proportion of transfected primary fetal astrocytes. We investigated the extent of HTV-1JR-FL. expression in fetal astrocytes and macrophages cultivated alone or together. Peak HIV-1 p24 antigen titres in supernatant fluids of macrophage cultures were increased with monocyte/macrophages from certain donors and were higher when macrophages were cocultivated with astrocytes. Structural HTV-1 gene (gp 41 and pot) products (protein and mRNA) were observed only in macrophages. Ten days after HTV-1JR-FL infection, astrocytes in a monoculture were stained negative or only weakly positive (1 - 2+) for Nef, whereas in a coculture up to 100% of astrocytes displayed Nef staining (up to 4+) in the cytoplasm. The streptavidine-biotine-peroxidase technique with certain monoclonal antibodies to Nef (Ovod et al, 1992) was specific for infected astrocytes. The intensity of Nef staining was higher in astrocytes cultivated with monocyte/macrophages from certain donors. In the coculture, tumor necrosis factor-α (TNF-α) was expressed in the astrocyte cytoplasm earlier after coinfection with HIV-1 and cytomegalovirus (CMV) compared to infection with HTV-1 alone. Interleukin-6 (IL-6) was secreted spontaneously and transiently in uninfected cocultures, but in a prolonged fashion following HTV-1 and HTV-1/CMV infections. The interactions between HTV-1- and CMV-infected macrophages and astrocytes lead to upregulation of TNF-a and IL-6 and enhancement of productive HIV-1 infection of macrophages and of ‘restricted’ HTV-1 infection of astrocytes with implications for the pathogenesis of AIDS dementia.
Article
Leukocyte recruitment from the blood circulation into tissue is essential for effective immune responses, and is, consequently, carefully regulated. In this article Yoshiya Tanaka and co-workers describe a model in which proteoglycans on the luminal surface of endothelium capture pro-adhesive cytokines. These cytokines provide the adhesion-inducing signal to particular leukocyte subsets which initiates their transmigration.
Article
The antigenic status of vascular endothelium from different sites of the normal adult and fetal human cardiovascular system was investigated. Tissues included aorta (n = 9), pulmonary artery (n = 8), coronary artery (n = 6), ventricle/atrium (n = greater than 10), lymph node (n = 2), fetal whole heart (n = 3), and umbilical cord (n = 7). Frozen sections were studied using monoclonal antibodies recognizing endothelial markers (EN4, vWf, Pal-E, and 44G4), vascular adhesion molecules (ICAM-1, ELAM, VCAM, and PECAM), the monocyte/endothelial marker (OKM5), and major histocompatibility complex (MHC) molecules (class I and class II). Results demonstrate that capillary endothelium is phenotypically different from endothelial cells (EC) lining large vessels. Capillary EC strongly express MHC classes I and II, ICAM, and OKM5, which are variably weak to undetectable on large vessels. In contrast, the large vessels strongly express vWf and appear to constitutively express ELAM-1. This suggests that the capillary EC may be more efficient at antigen presentation or more susceptible to immune attack in vivo. Interestingly, normal coronary arteries, unlike all other large vessels, express MHC class II and VCAM molecules. Future studies should concentrate on comparative functional studies between capillary, coronary, and large vessel EC.