ArticlePDF Available

Circulating human dendritic cells differentially express high levels of a 55-kD actin-bundling protein

Authors:

Abstract and Figures

This study was initiated to examine the differential expression of an evolutionary conserved human 55-kd actin-bundling (p55) protein that is induced in B lymphocytes by Epstein-Barr virus infection. Our study demonstrates that p55 is specifically expressed at constitutively high levels in human peripheral blood dendritic cells and lymph node (interdigitating) dendritic cells. Blood dendritic cells constitute a minority (< 2%) of all blood leukocytes but are a distinct population of potent antigen-presenting cells. Immunofluorescence microscopy with a monoclonal antibody specific for p55 showed that 87% of peripheral blood dendritic cells stained brightly in the cytoplasm and in the veiled cytoplasmic extensions. In contrast, monocytes, granulocytes, T cells, and B lymphocytes showed no expression of the p55 protein. Western blot analysis confirmed that only the dendritic cell component of peripheral blood expressed high levels of p55. Staining of human lymph node sections demonstrated selective expression of the p55 antigen by dendritic cells in the T-cell-dependent areas but not in the B cell follicles. p55 is likely to be involved in the organization of a specialized microfilament cytoskeleton in the dendritic cells, and the anti-p55 antibody should be useful for further characterization of this important population of antigen-presenting cells in clinical transplantation, HIV-1 pathogenesis, and autoimmune diseases.
Content may be subject to copyright.
American
journal
of
Patbology,
Vol.
148,
No.
2,
February
1996
Copyright
©)
Americani
Society
for
Investigative
Pathology
Circulating
Human
Dendritic
Cells
Differentially
Express
High
Levels
of
a
55-kd
Actin-Bundling
Protein
George
Mosialos,*
Mark
Birkenbach,t
Seyoum
Ayehunie,*
Fumio
Matsumura,§
Geraldine
S.
Pinkus,11
Elliott
Kieff,*
and
Erik
Langhoffl
From
the
Department
of
Microbiology
and
Molecular
Genetics
and
Medicine,*
Harvard
Medical
School,
the
Department
of
Human
Retrovirology,t
Dana-Farber
Cancer
Institute,
the
Department
of
Pathology,"1
Brigham
and
Women's
Hospital,
and
the
Renal
Unit,9
Massachusetts
General
Hospital,
Boston,
Massachusetts;
The
Mariorie
B.
Kovler
Viral
Oncology
Laboratories,t
The
University
of
Chicago,
Chicago,
Illinois;
and
the
Department
of
Molectular
Biology
and
Biochemistry,5
Rutgers
Universitv,
Piscataway,
New
Jersey
This
study
was
initiated
to
examine
the
differential
expression
of
an
evolutionary
conserved
human
55-kd
actin-bundling
(p55)
protein
that
is
induced
in
B
lymphocytes
by
Epstein-Barr
virus
infection.
Our
study
demonstrates
that
p55
is
specifically
expressed
at
constitutively
high
levels
in
human
peripheral
blood
dendritic
ceUls
and
lymph
node
(interdigitating)
dendritic
ceUls.
Blood
dendritic
cells
constitute
a
minority
(<2%)
of
aUl
blood
leu-
kocytes
but
are
a
distinct
population
of
potent
an-
tigen-presenting
cells.
Immunofluorescence
mi-
croscopy
with
a
monoclonal
antibody
specific
for
p55
showed
that
87%
ofperipheral
blood
dendritic
cels
stained
brightly
in
the
cytoplasm
and
in
the
veiled
cytoplasmic
extensions.
In
contrast,
mono-
cytes,
granulocytes,
T
ceUls,
and
B
lymphocytes
showed
no
expression
of
the
p55
protein.
Western
blot
analysis
confirmed
that
only
the
dendritic
ceUl
component
ofperipheral
blood
expressed
high
lev-
els
of
p55.
Staining
of
human
lymph
node
sections
demonstrated
selective
expression
of
the
p55
anti-
gen
by
dendritic
cells
in
the
T-ceUl-dependent
areas
but
not
in
the
B
cell
foUlicles.
p55
is
likely
to
be
involved
in
the
organization
of
a
specialized
mi-
crofilament
cytoskeleton
in
the
dendritic
cells,
and
the
anti-pSS
antibody
should
be
usefulforfurther
characterization
of
this
important
population
of
antigen-presenting
cells
in
clinical
transplantation,
HIV-1
pathogenesis,
and
autoimmune
diseases.
(Am
J
Pathol
1996
14&8593-600)
Blood
dendritic
cells
(DCs)
constitute
less
than
2%
of
peripheral
blood
leukocytes
and
are
highly
potent
antigen presenting
cells.1
In
lymphoid
organs,
circu-
lating
DCs
localize
to
the
T-cell-dependent
area
as
interdigitating
DCs
(IDCs).
DCs
are
15
to
20
,um
and
are
motile
with
minimal
phagocytic
activity.
DCs
ex-
press
high
levels
of
class
and
11
major
histocompat-
ibility
complex
proteins
and
are
bone-marrow-de-
rived
cells.1
2
A
different
type
of
cell
with
dendritic
morphology
localizes
in
the
B
cell
zones
(germinal
centers)
as
follicular
DCs
(FDCs).
FDCs
are
not
leu-
kocytes
and do
not
act
as
traditional
antigen-pre-
senting
cells
for
T
cells
but
are
involved
in
B
cell
proliferation
and
differentiation.3'4
The
study
of
human
blood
DCs
has
been
severely
restricted
by
lack
of
selective
markers
for
this
highly
specialized
subset
of
antigen-presenting
cells.
In
the
course
of
studying
the
tissue-restricted
expression
of
a
newly
characterized
human
actin-bundling
protein,
p55,
the
expression
of
which
is
highly
induced
by
Epstein-Barr
virus
infection,
we
discovered
that
p55
was
present
in
cells
of
dendritic
phenotype
in
the
T-cell-dependent
zones
of
the
lymph
nodes.
The
cDNA
for
p55
has
only
recently
been
cloned
and
has
been
characterized
as
an
actin-bundling
protein
originally
described
and
purified
from
HeLa
cells.5,6
A
monoclonal
antibody
against
p55
has
been
de-
rived
and
was
used
in
the
present
study
to
examine
p55
expression
in
human
leukocyte
subsets.
The
cells
expressing
p55
in
lymph
nodes
had
long
cyto-
plasmic
processes
characteristic
of
interdigitating
DCs.
As
these
cells
are
believed
to
be
derived
from
E.
L.
was
supported
by
National
Institutes
of
Health
grant
A128734.
Accepted
for
publication
October
19,
1995.
Address
reprint
requests
to
Dr.
Erik
Langhoff,
Massachusetts
General
Hospital,
Renal
Unit,
Boston,
MA
02114.
593
594
Mosialos
et
al
A/P
February
1996,
Vol.
148,
No.
2
Table
1.
55-kd
Protein
Expression
in
Subsets
of
Primary
Leukotes
from
a
Healthy
Donor
Cell
fraction
Cell
type
Anti-p55
Anti-DR
Anti-CD3
Anti-CD20
Anti-CD14/anti-CD11b
Anti-CD56
PBMCs
Granulocytes
Monocytes
T
cells
DC-enriched
fraction*
DC-depleted
fraction
(B
cell
enriched)
FACS-sorted
B
cells
FACS-sorted
DCs
0.9
<1
6
0
54
3
<1
96
87
(bright)
32
<1
71
2
80
34
>95
97
95
(bright)
51
<1
<1
95
8
24
9
<1
1
<1
6
18
<1
>95
<1
<1
ND,
not
determined.
*From
5
donors;
mean
=
50.4%,
SD
=
6.1%.
peripheral
blood
DCs,
we
set
out
to
investigate
p55
expression
in
peripheral
blood
DCs.
Materials
and
Methods
Cell
Fractionation
and
Cell
Cultures
Techniques
for
isolation
of
leukocyte
subsets
have
previously
been
described.7
Peripheral
blood
mononuclear
cells
(PBMCs)
were
isolated
by
Ficoll-
Hypaque
(Pharmacia,
Uppsala,
Sweden)
density
separation
from
blood
donor
buffy
coats
or
leuko-
paks
(fraction
1,
Table
1).
Granulocytes
were
iso-
lated
from
the
red
cell
pellet
of
the
Ficoll-Paque
gradients
after
lysis
of
the
red
cells
(fraction
2).
Next,
the
PBMC
fraction
was
separated
on
Percoll
density
gradients.
Monocytes
were
low
density
cells
and
were
depleted
of
contaminating
T
cells
by
rosetting
with
neuraminidase-treated
sheep
erythrocytes
(fraction
3).
T
cells
were
high
density
cells
positively
enriched
by
rosetting
with
sheep
erythrocytes
and
were
depleted
of
accessory
leukocytes
and
B
cells
by
nylon
wool
column
separation
(fraction
4).
This
procedure
yielded
>94%
pure
T
cells
as
determined
by
immunostaining.7
The
sheep-erythrocyte-rosette-
negative
cells
were
depleted
of
contaminating
monocytes
by
Fc
panning
of
monocytes
on
IgG-
coated
dishes.7
Low
density
cells
of
this
population
(enriched
for
DCs)
were
isolated
from
14.5%
metri-
zamide
(Sigma
Chemical
Co.,
St.
Louis,
MO)
gradi-
ents
at
500
x
g
for
12
minutes
at
room
temperature.
This
procedure
yielded
a
population
of
cells
that
was
estimated
to
be
50
to
70%
DCs
in
accordance
with
previous
studies7-9
(fraction
5).
The
high
density
cells
from
the
pellet
of
the
metrizamide
gradients
(depleted
of
DCs)
were
also
recovered.
This
fraction
is
enriched
in
B
cells
(fraction
6).
Purified
B
cells
(fraction
7)
were
obtained
by
fluorescein
isothiocya-
nate
(FITC)
FACS
sorting
for
CD19-positive
cells
of
fraction
6.
Purified
DCs
were
obtained
by
FACS
sort-
ing
by
positive
selection
for
large
cells
(high
side
scatter
and
high
forward
scatter).
More
than
95%
of
these
sorted
cells
had
on
the
hemocytometer
the
distinctive
phenotype
of
large
veiled
cells
and
were
strongly
positive
for
HLA-DR
antigen
(fraction
8).
Leukocyte
Cultures
The
function
of
monocytes
and
DCs
was
examined
by
comparing
their
antigen-presenting
functions
in
the
mixed
leukocyte
reaction
(MLR).
FACS-purified
blood
DCs
or
plastic-adherent
(Percoll-isolated)
monocytes
were
used
as
stimulator
cells
for
the
MLR
cultures
as
previously
described.10
The
stimulator
cells
were
irradiated
(3000
rads,
1
rad
=
0.01
Gy)
from
a
Cs127
source.
By
limiting
dilution,
graded
doses
of
stimulator
cells
(25,000
to
1,500
DCs
or
monocytes)
were
added
to
cultures
of
50,000
allo-
geneic
T
cells
in
round-bottom
96-well
plates
(Lim-
bro,
Flow
Laboratories,
Inc,
McLean,
VA)
with
growth
medium
of
RPMI
1640
and
10%
fetal
calf
serum.
The
MLR
cultures
were
cultured
for
5
days
before
addi-
tion
of
[3H]thymidine
(1
gCi/well;
DuPont-NEN,
Wilm-
ington,
DE)
for
20
hours.7'10
Phytohemagglutinin
(20
,Lg/ml;
Difco
Laborato-
ries,
Detroit,
Ml)
was
used
for
stimulation
of
T
cell
cultures
(fraction
3)
and
pokeweed
mitogen
(1:10
and
1:100;
Gibco,
Grand
Island,
NY)
was
used
for
stimulation
of
FACS-sorted
B
cells
(fraction
7)
for
5
days
before
harvest
of
the
cells
for
immunohisto-
chemistry.
1
1
Western
Blot
Analysis
The
presence
of
the
55-kd
actin-bundling
protein
was
analyzed
by
Western
blot
analysis.
Whole
cell
lysates
(4
x
105
primary
cells)
were
generated
by
boiling
the
cells
in
sodium
dodecyl
sulfate
(SDS)
1.
2.
3.
4.
5.
6.
7.
8.
23/ND
6/91
81/ND
6/ND
6/ND
12/ND
3/ND
<1/ND
2
<1
<1
<1
ND
<1
Leukocyte
Expression
of
Actin-Bundling
Protein
595
AJP
February
1996,
Vol.
148,
No.
2
loading
buffer
for
10
minutes
followed
by
centrifuga-
tion
to
remove
insoluble
material.
Whole
cell
isolates
(1
x
105
cell
equivalents)
were
analyzed
on
an
8.5%
SDS
polyacrylamide
gel
followed
by
electrophoretic
transfer
onto
nitrocellulose
membrane
and
immuno-
blotted
using
the
anti-p55
monoclonal
antibody
di-
luted
1:500
in
phosphate-buffered
saline
(PBS)
con-
taining
0.05%
Tween-20
and
3%
nonfat
dry
milk.
An
alkaline-phosphatase-conjugated
goat
anti-mouse
antibody
(Promega,
Madison,
WI)
was
used
at
1:5000
dilution
as
secondary
antibody
for
the
chro-
mogenic
detection
of
protein
bands
reactive
with
the
anti-p55
monoclonal
antibody.
Purified
55-kd
actin-
bundling
protein
from
HeLa
cells
was
run
in
parallel
control
lanes.5
Monoclonal
Antibodies
and
Immunohistochemistry
A
monoclonal
antibody
to
the
55-kd
protein
was
raised
as
previously
described.12
Female
BALB/57
mice
were
immunized
intraperitoneally
with
20
,ug
of
purified
55-kd
protein
in
complete
Freund's
adjuvant
and
after
1
month
with
20
,ug
of
55-kd
protein
in
incomplete
Freund's
adjuvant.
The
mice
were
re-
boosted
12
times
before
harvest
of
the
spleens
for
fusion
with
the
mouse
myeloma
cell
line
NS-1
and
enzyme-linked
immunosorbent
assays
were
used
to
screen
for
positive
clones.12
One
clone,
K-2
(IgG1),
of
three
clones
reactive
with
the
55-kd
protein,
was
used
for
the
present
studies.
Culture
supernatants
of
the
K-2
hybridoma
were
used
directly
as
an
antibody
source
for
the
staining
reactions.
Ascites
fluid
was
also
obtained
by
intraperitoneal
injection
of
the
K-2
hybridoma
into
BALB/c
mice
(Hybridoma
Core
Fa-
cility,
Dana-Farber
Cancer
Institute).
Both
hybridoma
supernatant
(undiluted)
and
ascites
(diluted
1:500
to
1:1000)
were
used
in
the
studies
and
gave
consis-
tent
results.
Antibodies
against
leukocyte
differentiation
anti-
gens
were
used
according
to
the
manufacturer's
instructions
unless
otherwise
specified:
anti-HLA-DR
(9.3F10,
IgG2,,
American
Type
Culture
Collection
(ATCC),
Rockville,
MD,
or
Anti-HLA-DR,
lgG21,
Bec-
ton
Dickinson
(BD),
San
Jose,
CA),
anti-CD3
(leu
4,
IgG1,
BD),
anti-CD19/CD20
(leu
12,
IgG1,
BD/Leu
16,
IgG1,
BD),
anti-CD14
IgG1),
anti-CD14
(3C-
10,lgG2a,
ATCC,
or
leu-M3,
IgG1,
BD),
anti-CD11b
(OKM1,
IgG1,
ATCC)
or
anti-CD56
(leu
19,
IgG1,
BD),
anti-CD1a
(OKT6,
IgG1,
ATCC).
The
antibody
specific
to
the
Epstein-Barr
virus
nuclear
protein
2
(EBNA
2)
was
PE2
(IgG1,
Young
et
al13).
For
immunohistochemical
tissue
localization
of
p55
or
CD1a,
acetone-fixed
cryostat
sections
of
the
reactive
lymph
node
were
incubated
with
monoclo-
nal
antibodies
for
1
hour.
Slides
were
washed
with
Tris-buffered
saline
and
then
incubated
sequentially
with
rabbit
anti-mouse
immunoglobulin
antibodies
(Dako
Corp.,
Carpinteria,
CA;
1:40
dilution,
using
0.1
mol/L
Tris/HCL
buffer,
pH
7.6,
supplemented
with
4%
human
AB
serum
as
diluent)
and
calf
alkaline
phosphatase
anti-calf
alkaline
phosphatase
immune
complexes
(APAAP;
Dako
Corp;
1:50
dilution).
Anti-
body
localization
was
effected
using
an
alkaline
phosphatase
reaction
with
naphthol
AS-MX
phos-
phate
(Sigma)
as
substrate
and
Fast
Red
TR
salt
(Sigma)
as
chromogen,
as
described
by
Cordell
et
al.14
For
negative
controls,
isotype-specific
mouse
immunoglobulin
or
Tris
buffer,
respectively,
were
substituted
for
the
primary
antibody
in
sequential
sections.
Cytocentrifuge
preparations
of
peripheral
blood
DCs
were
processed
as
positive
controls.
Cy-
tofluorography
of
cell
isolates
was
performed
as
pre-
viously
described.7
Cytospin
centrifuge
preparations
were
made
of
all
isolated
leukocyte
subsets
(20,000
cells
per
slide;
Shandon,
Southern
Instruments,
Pittsburgh,
PA).
Procedures
for
cytospin
immunofluorescence
have
previously
been
described.815
Briefly,
cytocentri-
fuge
preparations
were
fixed
for
30
minutes
in
ice-
cold
methanol.
After
drying,
the
slides
were
stored
at
-70°C
until
use.
Immunofluorescence
localization
of
the
55-kd
protein
and
leukocyte
differentiation
anti-
gens
was
done
on
cytocentrifuge
slides
presoaked
in
PBS
with
1%
human
serum
and
1%
fetal
calf
serum.
The
slides
were
then
sequentially
incubated
with
titered
primary
antibody
reagents
followed
by
FITC-conjugated
secondary
goat
anti-mouse
anti-
body
(1:200;
Sigma).
For
negative
controls,
isotype-
specific
mouse
immunoglobulin
or
PBS
with
1%
fetal
calf
serum,
respectively,
were
substituted
for
the
primary
antibody
in
sequential
cytospin
prepara-
tions.
For
each
antibody
used,
the
frequency
(percent-
age)
of
reactive
cells
was
counted
independently
by
two
investigators
using
results
of
cell
counts
from
at
least
five
fields
or
more
than
250
cells.
Results
The
reactivity
of
the
anti-p55
antibody
was
examined
by
immunofluorescence
staining
of
cultures
of
PB-
MCs
and
purified
subsets
of
granulocytes,
mono-
cytes,
T
cells,
B
cells,
enriched
DCs,
and
FACS-
purified
DCs.
Table
1
presents
results
of
analysis
of
sequential
steps
of
leukocyte
purification
from
a
healthy
donor.
A
panel
of
monoclonal
antibodies
596
Mosialos
et
al
AJP
Febrary
1996,
Vol.
148,
No.
2
*.4
..,.-s
e.
....
..^....X
.::..........
:4
...,.
.:xeX
......
a
c
Figure
1.
Jnmuno/fiuorescence
FITC
staining
of
bulk
PBMCs
ancd
FACS-ptinfied
blood
DCs.
a:
Phase
contrast
coittrol
(X
200)
of
PBMCs.
The
arrow
shous
a
cell
that
expresses
the
p55
antigen
by
immunofluorescence
(b).
b:
p55
FITCstaining
of
the
PBMCfield
shoun
in
a.
c:
Phase
contrast
conttrol
(X
630)
of
FACS-purified
blood
DCs.
d:
p55
FITC
stainzitng
of
FACS-purified
DC,.
specific
for
leukocyte
differentiation
antigens
was
used
in
parallel
with
the
anti-p55
antibody.
Among
unseparated
PBMCs,
0.9%
of
the
cells
were
found
to
be
brightly
positive
by
immunofluores-
cence
microscopy
for
p55
(Table
1,
fraction
1;
Figure
1,
a
and
b).
In
a
second
experiment,
<2%
of
PBMCs
were
positive
for
p55.
When
isolated
leukocyte
sub-
sets
were
stained
for
p55,
most
of
the
p55-positive
cells
were
in
the
metrizamide-separated
fraction
of
enriched
DCs
(fraction
5).
In
this
fraction,
54%
of
the
cells
were
positive
for
p55,
but
this
preparation
also
contains
some
cell
contaminants
from
other
leuko-
cyte
subsets.
Similar
yields
of
DCs
have
been
ob-
tained
in
repeated
experiments
(mean
=
50.4,
SD
=
6.1,
n
=
5).
To
eliminate
cell
contaminants
from
other
leukocyte
subsets,
DCs
were
further
purified
by
FACS
sorting
for
large
cells;
87%
of
this
population
of
purified
DCs
stained
strongly
for
p55
(Table
1,
frac-
tion
8,
Figure
1,
c
and
d).
When
both
bright
and
medium
intensity
cells
were
counted,
96%
of
the
cells
were
positive
for
p55.
As
expected,
95%
of
these
highly
purified
DCs
were
also
strongly
positive
for
HLA-DR
(not
shown).
A
second
experiment
of
FACS
purification
of
DCs
yielded
95%
cells
positive
(bright
and
medium)
for
p55
and
less
than
1
%
reac-
tive
with
a
cocktail
of
anti-CD14,
anti-CD3,
and
anti-
CD57.
In
both
experiments,
the
expression
of
p55
as
measured
by
immunofluorescence
was
comparable
to
that
of
HLA-DR
expression
by
DCs
and
p55
by
Epstein-Barr-virus-infected
cell
lines.
Cytofluorogra-
phy
was
performed
in
two
experiments
and
showed
no
surface
expression
of
p55
by
isolated
DCs
or
T
cells.
p55
localized
diffusely
to
the
cytoplasm
and
was
also
evident
along
dendritic
projections.
In
the
p55-
positive
cells,
the
p55
staining
coincided
with
actin
staining
(not
shown).
p55
staining
was
specific
as
isotype-matched
control
antibodies
(anti-CD3,
anti-
CD20,
and
anti-CD56;
Table
1)
failed
to
stain
DCs.
Monocytes,
granulocytes,
T
cells,
or
B
cells
(frac-
tions
2,
3,
4,
6,
and
7)
were
not
positive
for
p55.
In
two
sets
of
experiments,
phytohemagglutinin-stimu-
lated
cultures
of
T
cells
and
pokeweed-mitogen-
stimulated
cultures
of
FACS-sorted
B
cells
yielded
the
same
low
frequency
(<1%)
of
p55-positive
cells
after
5
days
of
stimulation
as
that
of
unstimulated
cultures
(<1%).
Figure
2
shows
Western
blot
analysis
of
p55
ex-
pression
in
isolated
leukocyte
subsets.
The
Western
blot
analysis
confirmed
the
results
of
the
immunoflu-
orescence
staining.
Thus,
by
Western
blot
analysis,
p55
was
found
to
be
abundant
in
the
DC
fraction
but
Leukocyte
Expression
of
Actin-Bundling
Protein
597
A/P
Februiary
1996,
Vol.
148,
No.
2
1
2 3
4
5
-97
kD
-
66
kD
-45
kD
Figure
2.
Western
blot
analysis
ofpiS
expression
in
v'arious
primnary
lenktoc
te
sub.sets.
The
following
samples
uere
anialyzed:
lane
1,
whole
ccell
extracftromn
1
x
105
DCs;
lane
2,
wbole
cell
extractfromn
I
X
10J
mnoniocytes;
lane
3.
whole
cell
extractifomn
1
x
10
enrliched
B
cells.
lane
4,
nhole
cell
extract
from
I
x
10"
bulk
T
cells;
lane
5,
purified
55-kd
protein
from
HeLa
cells.
The
arrow
indicates
the
position
()/'the
55-kd
actini-btonidlinig
protein.
was
not
detectable
in
other
examined
leukocyte
sub-
sets.
The
functional
capacity,
as
antigen-presenting
cells,
of
the
FACS-sorted
95%
HLA-DR-positive/87%
p55-positive
DCs
was
examined
(fraction
8).
For
these
studies,
the
mixed
lymphocyte
stimulatory
ac-
tivity
(MLR)
of
DCs
was
compared
with
that
of
iso-
lated
monocytes
(Table
1,
fraction
3).
Figure
3
shows
the
results
of
the
MLR
studies
using
limiting
dilutions
of
DCs
or
monocytes
as
stimulator
cells
and
alloge-
neic
T
cells
as
responder
cells.
By
comparison,
the
half-maximal
stimulatory
capacity
of
DCs
was
20-fold
higher
than
that
of
monocytes
(Figure
3).
In
cryostat
sections
of
a
lymph
node,
the
reactivity
for
p55
was
localized
to
mononuclear
cells
in
inter-
follicular
areas
(T
cell
zones)
of
the
node
(Figure
4a).
The
immunoreactive
cells
revealed
a
dendritic
ap-
pearance,
consistent
with
IDCs,
and
exhibited
strong,
cytoplasmic
staining
for
the
p55
(Figure
4b).
The
p55-positive
cells
in
the
T-cell-dependent
zone
constituted
<3%
of
the
cell
population
in
tissue
sec-
tions.
Lymphoid
cells
in
the
T
and
B
cell
zones
were
uniformly
nonreactive.
Similar
results
have
been
ob-
tained
from
lymph
node
specimens
from
four
differ-
ent
sources.
Follicular
DCs
of
B
cell
zones
were
nonreactive,
although
focally,
weak
staining
was
noted
for
occasional
cells.
Studies
of
sequential
sec-
30
Q
C.)
0-
co
C)
CD
20
E
10
-ce
I-
2
1.5
x
103
3
x
103
6
x
103
12.5
x
103
25
x
103
Stimulator
Cells/Well
Figure
3.
Mixced
leukocyte
reaction.
The
MLR
stinmlatory'
capacity
qf
FACS-enriched
DCs
(-)
and
enicbhed
monocytes
(0)
uere
co-culltulred
wvitb
50,
000
allogeneic
T
cells.
As
stimullator
cells
1,
500
to
25,
000
cells
were
used
in
the
experiment.
Day
5
PHithymidine
incorporation
is
sbo'wn
on
the
ordinate.
Number
of
added
(y-irradiated)
stimnlator
DCs
or
inoniocytes
is
shon'n
on?
the
abscissa.
Day
S
13Hithymnidine
incotporation
of
50,000
Tcell.s
alone
was
120
cpmt?
(niot
shown'i).
tions
using
monoclonal
antibody
(OKT6
(CDla)),
re-
vealed
only
small
numbers
of
reactive
cells
in
the
same
distribution
as
the
p55-positive
cells
(Figure
4c).
In
preliminary
studies,
tissue
thymic
DCs
have
also
been
shown
to
be
positive
for
p55
with
predom-
inant
localization
to
the
medulla.
Few
cells
in
kidney
sections
have
staining
for
p55
at
a
frequency
com-
parable
to
cells
expressing
HLA-DR.
Work
is
cur-
rently
in
progress
to
further
characterize
those
p55-
positive
cells.
Discussion
The
present
results
demonstrate
that
among
periph-
eral
blood
cells,
DCs
express
high
levels
of
p55
whereas
monocytes,
T
lymphocytes,
B
lymphocytes,
and
granulocytes
do
not
express
p55.
In
PBMC
cul-
tures
(Table
1),
0.9%
of
the
cells
were
p55
positive,
which
is
in
agreement
with
the
expected
frequency
of
DCs
reported
in
other
studies.1'2
Thus,
the
present
results
suggest
that
p55
expression
is
a
useful
marker
for
identifying
and
potentially
for
quantitating
the
number
of
DCs
in
peripheral
blood
or
in
leuko-
cyte
subsets.
Although
virtually
all
FACS-isolated
DCs
expressed
at
least
moderate
levels
of
p55
(95%
of
the
FACS-sorted
cells
versus
97%
expressing
high
levels
of
HLA-DR),
87%
of
the
FACS-sorted
cells
displayed
high
levels
of
p55.
DCs
that
express
only
a
moderate
level
of
p55
may
constitute
a
distinct
fraction
of
peripheral
blood
DCs
at
a
different
stage
4
-
=00.
598
Mosialos
et
al
AJP
February
1996,
Vol.
148,
No.
2
Figure
4.
Lymph
node,
cryostat
section.
a:
At
lonl!
miagnification
(X200),
frequent
mononuclear
cells
immunoreactiveforpSi
are
observed
in
the
interfollicular
area
of
the
node.
Lymphoid
cells
are
nonreactive.
b:
Higher
miagtnificationi
demonstrates
strong
cytoplasmic
reactivity
for
p55
in
the
population
of
DCs
in
the
interfollictular
regioni
of
the
node.
Note
the
comnplex
elonigated
cellprocesses
of
the
immunoreactive
cells.
c:
In
a
sequential
section
of
the
reactive
niode,
only
small
numbers
of
mononuiclear
cells,
morphbologically
consistent
with
DCs,
are
reactivefor
OKT6(
CDla).
These
cells
are
also
localized
to
inteifollicular
areas.
Immunoalkaline
phosphatase
technique;
methyl
greeni
counterstain.
M1,
niantle
zone:
G,
germninial
center.
of
differentiation
or
of
the
cell
cycle.
Mosialos
et
al5
have
previously
shown
that
p55
is
undetectable
by
Western
blot
analysis
in
two
actively
proliferating
Epstein-Barr-virus-negative
B
lymphoma
cell
lines
(BJAB
and
BL41)
and
is
also
undetectable
by
North-
ern
blot
analyis
in
three
Epstein-Barr-virus-negative
B
lymphoma
cell
lines
(loukes,
BL30,
and
BL41)
and
two
T
cell
lines
(Jurkat
and
MOLT4).
These
results
further
support
the
selective
expression
of
p55,
al-
though
we
cannot
exclude
the
possibility
that
a
rare
B
cell
or
T
cell
population
(less
than
1%
of
the
total
B
and
T
cell
population)
may
express
p55.
In
phytohe-
magglutinin-activated
PBMC
cultures
containing
DCs,
a
transient
up-regulation
of
p55
RNA
(14
to
48
hours)
has
been
reported.16
The
results
also
show
that
the
expression
of
p55
in
lymph
node
sections
is
restricted
to
cells
resembling
IDCs
and
is
not
found
expressed
in
the
follicles.
This
is
in
agreement
with
the
current
theory
that
IDCs
are
derived
from
a
cell
lineage
different
from
FDCs.34
IDCs
in
the
lymph
node
are
thought
to
be
derived
from
peripheral
blood
DCs
migrating
to
the
lymphoid
tissues.
17,18
A
small
number
of
CDla-positive
DCs
was
found
in
lymph
node
sections
in
the
T
cell
zones.
CDla
is
thought
to
be
an
early
maturation
marker
present
on
bone-marrow-derived
DCs.
The
low
frequency
of
CDla-positive
DCs
could
be
explained
by
a
small
number
of
immature
DCs
in
the
lymphoid
tissues
or
a
loss
of
the
CDla
antigen
upon
maturation
in
periph-
eral
blood
and
in
lymph
nodes.19
20
The
extent
of
the
reactivity
with
cell
subsets
in
other
tissues
is
pres-
ently
under
investigation.
Both
in
vitro
studies
and
tissue
studies
have
not
demonstrated
reactivity
with
activated
B
cells
in
culture
or
staining
with
B
cell
follicles
that
contain
activated
B
cells.
It
is
possible
that
the
p55
antigen
is
also
expressed
in
immature
bone-marrow-derived
DCs
and
Langerhans
cells.
Initial
studies
(not
shown)
have
indicated
that
Lang-
erhans
cells
express
the
p55
antigen,
but
additional
studies
are
in
progress
to
asses
the
extent
of
p55
expression
in
immature
DCs.
Several
studies
of
tis-
sue
sections
from
thymus
(two
samples),
lymph
node
(three
samples),
and
tonsil
(one
sample)
have
shown
that
cells
of
dendritic
morphology
and
reac-
tive
with
the
p55
antibody
consistently
localize
in
the
T-cell-dependent
areas
of
the
lymph
node
and
tonsil
as
well
as
in
the
medulla
of
the
thymus.
In
kidney
sections
(four
samples),
occasional
cells
of
dendritic
morphology
in
the
interstitium
stain
for
the
p55
anti-
gen.
Proliferating
cells
in
histiocytosis
X
are
thought
to
be
derived
from
cells
of
the
dendritic
lineage.21'22
Preliminary
data
(Birkenbach
et
al,
personal
commu-
nication)
have
shown
that
cells
from
histiocytosis
X
lesions
express
the
p55
antigen.
p55
is
one
of
the
few
markers
for
human
DCs.
Recent
work
has
described
expression
of
the
HB-15
antigen
by
DCs
of
the
blood
and
thymus.23
However,
the
HB-15
antigen
is
also
expressed
by
FDCs.
Fur-
thermore,
the
HB-15
antigen
is
expressed
by
acti-
vated
T
cells.23
The
S-100
antigen
has
a
wide
tissue
distribution
and
is
expressed
in
many
cell
types
of
different
somatic
origin
including
DCs.24
I&
Leukocyte
Expression
of
Actin-Bundling
Protein
599
AJP
February
1996,
Vol.
148,
No.
2
The
expression
of
p55
by
DCs
adds
to
the
com-
plexity
of
the
differentiation-specific
expression
of
this
particular
actin-bundling
protein.
Actin
bundling
proteins
regulate
rearrangements
of
the
cytoskeleton
or
interaction
between
the
cytoskeleton
and
mem-
branes
in
response
to
extra-
or
intracellular
sig-
nals.25
Actin-bundling
proteins
direct
the
three-
dimensional
polymerization
of
actin
filaments
in
mi-
grating
cells.
Related
to
cell
motility
is
also the
ability
to
process
and
present
antigen.25
Thus,
the
migra-
tory
nature
and
unique
antigen-presenting
proper-
ties
of
DCs
suggest
a
critical
role
of
p55
in
this
specialized
subset
of
leukocytes.
p55
is
highly
con-
served
from
sea
urchin
through
Drosophila
to
mam-
malian
species.5
In
Drosophila,
mutations
in
p55
re-
sult
in
a
singed
hair
phenotype
and
sterility.
In
humans,
p55
expression
is
found
also
in
the
den-
drites
of
specific
neurons
in
the
central
nervous
sys-
tem.
In
rats,
a
high
level
of
expression
has
also
been
demonstrated
in
neural
cells
in
vitro
and
in
neural
growth
cones.5
Migration
of
axons
during
the
devel-
opment
of
the
fetal
nervous
system
is
functionally
related
to
the
process
of
cell
migration.25
In
vivo
engagement
of
DCs
in
disease
is
sug-
gested
from
studies
in
rodents,
but
studies
in
hu-
mans
have
been
limited
by
the
lack
of
a
strongly
reactive
and
specific
marker
for
these
specialized
cells.
Of
immediate
clinical
interest
is
the
potential
use
of
the
p55
protein
as
a
marker
for
the
possible
engagement
of
dendritic
leukocytes
in
organ
trans-
plantation,
HIV-1
pathogenesis,
and
autoimmunity.
Acknowledgments
We
thank
A.
M.
Bruzzese
for
technical
assistance.
References
1.
Steinman
RM:
The
dendritic
cell
system
and
its
role
in
immunogenicity.
Annu
Rev
Immunol
1991,
9:271-296
2.
Steinman
RM,
Cohn
ZA:
Identification
of
a
novel
cell
type
in
peripheral
lymphoid
organs
of
mice.
J
Exp
Med
1974,
139:380-397
3.
Schreiver
F,
Nadler
LM:
The
central
role
of
follicular
dendritic
cells
in
lymphoid
tissues.
Adv
Immunol
1992,
51:243-283
4.
Klaus
GGB,
Humphrey
JH,
Kunki
A,
Dongworth
DW:
The
follicular
dendritic
cell:
its
role
in
antigen
presen-
tation
in
the
generation
of
immunological
memory.
Im-
munol
Rev
1980,
53:243-283
5.
Mosialos
G,
Yamashiro
S,
Baughman
R,
Matsudaira
R,
Matsumura
F,
Vara
L,
Kief
E,
Birkenbach
M:
Epstein-
Barr
virus
infection
induces
expression
in
B
lympho-
cytes
of
a
novel
gene
encoding
an
evolutionarily
con-
served
55-kd
actin-bundling
protein.
J
Virol
1994,
68:
7320-7324
6.
Yamashiro-Matsumura
S,
Matsumura
F:
Purification
and
characterization
of
an
F-actin-bundling
55-kilodal-
ton
protein
from
HeLa
cells.
J
Biol
Chem
1985,
260:
5087-5097
7.
Langhoff
E,
Steinman
RM:
Clonal
expansion
of
human
T
lymphocytes
isolated
by
dendritic
cells.
J
Exp
Med
1989,
169:315-320
8.
Langhoff
E,
Terwilliger
EF,
Bos
HJ,
Kalland
KH,
Poznansky
MC,
Bacon
OML,
Haseltine
WA:
Replication
of
human
immunodeficiency
virus
type
1
in
primary
dendritic
cells.
Proc
Natl
Acad
Sci
USA
1990,
88:7998-
8002
9.
Langhoff
E,
Kalland
KH,
Haseltine
WA:
Early
molecular
replication
of
human
immunodeficiency
virus
type
1
in
cultured-blood-derived
T
helper
dendritic
cells.
J
Clin
Invest
1993,
91
:2721-2726
10.
Langhoff
E,
Jakobsen
BK,
Platz
P,
Ryder
LP,
Sveigaard
A:
The
impact
of
low
donor-specific
MLR
versus
HLA-DR
compatibility
on
kidney
graft
survival.
Trans-
plantation
1985,
39:18-21
11.
Langhoff
E,
Ladefoged
J,
Dickmeiss
E:
The
immuno-
suppressive
potency
of
various
steroids
on
peripheral
lymphocytes,
T
cells,
NK
and
K
cells.
Int
J
Immuno-
pharmacol
1985,
7:483-489
12.
Yamashiro-Matsumura
S,
Matsumura
F:
Intracellular
lo-
calization
of
the
55-kd
actin-bundling
protein
in
cultured
cells:
spatial
relationships
with
actin,
a-actinin,
tropomy-
osin,
and
fimbrin.
J
Cell
Biol
1986,
103:631-640
13.
Young
L,
Alfieri
C,
Hennessy
K,
Evans
H,
O'Hara
C,
Anderson
KC,
Ritz
J,
Shapiro
RS,
Rickinson
A,
Kieff
E,
Cohen
JI:
Expression
of
Epstein-Barr
virus
transforma-
tion-associated
genes
in
tissues
of
patients
with
EBV
lymphoproliferative
disease.
N
EngI
J
Med
1989,
321:
1080-1085
14.
Cordell
JL,
Falini
B,
Erber
WN,
Ghosh
AK,
Abudulaziz
Z,
MacDonald
S,
Pulford
KAF,
Stein
H,
Mason
DY:
lmmunoenzymatic
labeling
of
monoclonal
antibodies
using
immune
complexes
of
alkaline
phosphatase
and
monoclonal
anti-alkaline
phosphatase
(APAAP
com-
plexes).
J
Histochem
Cytochem
1984,
32:219-229
15.
Kalland
KH,
Szilvay
AM,
Langhoff
E,
Haukeness
G:
Subcellular
distribution
of
human
immunodeficiency
vi-
rus
type
1
Rev
and
colocalization
of
Rev
with
RNA
splicing
factors
in
a
speckled
pattern
on
the
nucleo-
plasm.
J
Virol
1994,
68:1475-1485
16.
Duh
F-H,
Latif
F,
Weng
Y,
Geil
L,
Modi
W,
Stackhouse
T,
Matsumura
F,
Duan
DR,
Linehan
WM,
Lerman
Ml,
Gnarra
JR:
cDNA
cloning
and
expression
of
the
human
homolog
of
the
sea
urchin
fascin
and
Drosophila
singed
genes
which
encode
an
actin-bundling
protein.
DNA
Cell
Biol
1994,
13:821-827
17.
Macatonia
SE,
Edwards
AJ,
Knight
SC:
Dendritic
cells
and
the
initiation
of
contact
sensitivity
to
fluorescein
isothiocyanate.
Immunology
1986,
59:509-514
18.
Larsen
C,
Steinman
R,
Witmer-Pack
M,
Hankins
D,
Morris
P,
Austyn
J:
Skin
migration
and
maturation
of
600
Mosialos
et
al
AJP
February
1996,
Vol.
148,
No.
2
Langerhans
cells
in
skin
transplants
and
explants.
J
Exp
Med
1990,
172:1483-1493
19.
Schuler
G,
Steinman
RM:
Murine
epidermal
Langer-
hans
cells
mature
into
potent
immunostimulatory
den-
dritic
cells
in
vitro.
J
Exp
Med
1985,
161
:526-546
20.
Romani
N,
Lenz,
A,
Glassel
H,
Stbssel
H,
Stanzl
U,
Majdic
0,
Fritsch
P,
Schuler
G:
Cultured
human
Langerhans
cells
resemble
lymphoid
dendritic
cells
in
phenotype
and
function.
J
Invest
Dermatol
1989,
93:600-609
21.
Beckstead
JH,
Wood
GS,
Turner
RR:
Histiocytosis
X
and
Langerhans
cells.
Hum
Pathol
1984,
15:826-833
22.
Willman
CH,
Busque
L,
Griffith
BB,
Blaise
MS,
Favara
E,
McGlain
KL,
Duncan
MH,
Gilliland
DG.
Langerhans'-
cell
histiocytosis
(histiocytosis
X):
a
clonal
proliferative
disease.
N
Engl
J
Med
1994,
331:154-157
23.
Zhou
L-J,
Scharting
R,
Smith
HM,
Tedder
TF:
A
novel
cell-surface
molecule
expressed
by
human
interdigitat-
ing
reticulum
cells,
Langerhans
cells
and
activated
lymphocytes
is
a
new
member
of
this
immunoglobulin
superfamily.
J
Immunol
1992,
149:735-742
24.
Vanstapel
M-J,
Gatter
KC,
de
Wolfe-Peeters
C,
Manson
DY,
Desmet
VD:
New
sites of
human
S-100
immunore-
activity
detected
with
monoclonal
antibodies.
Am
J
Clin
Pathol
1986,
85:160
25.
Stossel
TS:
On
the
crawling
of
animal
cells.
Science
1993,
260:1086-1094
... Indeed, we have previously reported a critical role for fascin in promoting breast cancer cell adhesion (Barnawi et al., 2019), invasion, and metastasis (Al-Alwan et al., 2011). In healthy tissues, fascin has restricted expression in selective cells such as dendritic cells, endothelial cells, neurons, especially near the dendritic protrusions (Duh et al., 1994;Mosialos et al., 1994Mosialos et al., , 1996Pinkus et al., 1997). While fascin is aberrantly expressed in various transformed epithelial cells, including the breast (Hashimoto et al., 2005), its expression in the normal breast has not been investigated before. ...
... Furthermore, qRT-PCR analysis of fascin RNA level in sorted mammary cell subsets reinforced its predominant expression in the stromal fraction ( Supplementary Fig. 3E). In agreement with previous results (Al-Alwan et al., 2001b;Hashimoto et al., 2011;Jayo et al., 2012;Mosialos et al., 1996), multicolor analysis of single mammary cells using flow cytometry showed fascin expression in fibroblasts, endothelial cells and immune cells (dendritic cells) (Supplementary Figurer 3F) of the stromal population. Altogether, immunohistochemistry and FACS results showed no obvious effect of fascin loss on mammary gland architecture or cellular composition of the three mammary gland fractions. ...
... Fascin was primarily observed in the stromal fraction of the mammary gland, particularly in fibroblasts, endothelial and immune cells (dendritic cells) consistent with previous reports (Hashimoto et al., 2011;Jayo et al., 2012). In addition, fascin expression has been reported in other organs/tissues such as central nervous system, spleen and mesenchymal of the placenta (Duh et al., 1994;Mosialos et al., 1996;Pinkus et al., 1997;Zhang et al., 2008). The use of whole body knockout model to assess the effect of fascin loss on lactogenesis pose a limitation in the current study, thus factors that are directly or indirectly impaired in the process of lactation can be explored in future studies. ...
Article
Full-text available
Fascin expression has commonly been observed in certain subtypes of breast cancer, where its expression is associated with poor clinical outcome. However, its role in normal mammary gland development has not been elucidated. Here, we used a fascin knockout mouse model to assess its role in normal mammary gland morphogenesis and lactation. Fascin knockout was not embryonically lethal, and its effect on the litter size or condition at birth was minimal. However, litter survival until the weaning stage significantly depended on fascin expression solely in the nursing dams. Accordingly, pups that nursed from fascin−/− dams had smaller milk spots in their abdomen, suggesting a lactation defect in the nursing dams. Mammary gland whole-mounts of pregnant and lactating fascin−/− mice showed significantly reduced side branching and alveologenesis. Despite a typical composition of basal, luminal, and stromal subsets of mammary cells and normal ductal architecture of myoepithelial and luminal layers, the percentage of alveolar progenitors (ALDH⁺) in fascin−/− epithelial fraction was significantly reduced. Further in-depth analyses of fascin−/− mammary glands showed a significant reduction in the expression of Elf5, the master regulator of alveologenesis, and a decrease in the activity of its downstream target p-STAT5. In agreement, there was a significant reduction in the expression of the milk proteins, whey acidic protein (WAP), and β-casein in fascin−/− mammary glands. Collectively, our data demonstrate, for the first time, the physiological role of fascin in normal mammary gland lactogenesis, an addition that could reveal its contribution to breast cancer initiation and progression.
... 19 During embryonic development and fetal maturity, fascin-1 is prominently expressed in the nervous system, including the dendritic cells, and in the microvascular endothelial cells, gastrointestinal tract, and mesenchymal tissue. [20][21][22][23][24][25] These cells are similar in terms of experiencing significant cell migration, indicating that fascin-1 fulfills an important role in cell migration. Moreover, fascin-1 expression is highly upregulated in cytotrophoblast cells (CTBs), promoting the proliferation of CTBs and, thus, the formation of the placenta in early gestation. ...
... [33][34][35] Fascin-1 expression is highly upregulated in mature dendritic cells, thus facilitating antigen presentation to T cells and participation in the immune response. 20,36 Within mature dendritic cells, fascin-1 can promote dendritic cell migration into lymph nodes by promoting podosome disassembly and increasing membrane protrusions. 36 Moreover, fascin-1 is involved in cell migration by combining with the microtubule cytoskeleton. ...
Article
Full-text available
Human cancer statistics show that an increased incidence of urologic cancers such as bladder cancer, prostate cancer, and renal cell carcinoma. Due to the lack of early markers and effective therapeutic targets, their prognosis is poor. Fascin-1 is an actin-binding protein, which functions in the formation of cell protrusions by cross-linking with actin filaments. Studies have found that fascin-1 expression is elevated in most human cancers and is related to outcomes such as neoplasm metastasis, reduced survival, and increased aggressiveness. Fascin-1 has been considered as a potential therapeutic target for urologic cancers, but there is no comprehensive review to evaluate these studies. This review aimed to provide an enhanced literature review, outline, and summarize the mechanism of fascin-1 in urologic cancers and discuss the therapeutic potential of fascin-1 and the possibility of its use as a potential marker. We also focused on the correlation between the overexpression of fascin-1 and clinicopathological parameters. Mechanistically, fascin-1 is regulated by several regulators and signaling pathways (such as long noncoding RNA, microRNA, c-Jun N-terminal kinase, and extracellular regulated protein kinases). The overexpression of fascin-1 is related to clinicopathologic parameters such as pathological stage, bone or lymph node metastasis, and reduced disease-free survival. Several fascin-1 inhibitors (G2, NP-G2-044) have been evaluated in vitro and in preclinical models. The study proved the promising potential of fascin-1 as a newly developing biomarker and a potential therapeutic target that needs further investigation. The data also highlight the inadequacy of fascin-1 to serve as a novel biomarker for prostate cancer.
... Multiple cell types within the cardiovascular system express FSCN1, including dendritic cells, B lymphocytes, T lymphocytes, macrophages, neutrophils, platelets, vessel wall endothelial cells, smooth muscle cells, and fibroblasts [48]. In normal human peripheral blood, FSCN1 expression is restricted to dendritic cells, which play a primary role in the initiation of acquired immune responses ( Figure 2) [49][50][51]. ...
Article
Full-text available
Fascin actin-bundling protein 1 (FSCN1) is an actin-bundling protein that is capable of inducing membrane protrusions and plays critical roles in cell migration, motility, adhesion, and other cellular interactions. FSCN1 also plays a role in forming and stabilizing filopodia or microspikes, which assist during cell migration. Furthermore, FSCN1 is a downstream target of several microRNAs and participates in various biological processes, such as epithelial-to-mesenchymal transition and autophagy, which regulate the invasion and migration ability of cells in various cancers. Increased FSCN1 levels have been associated with enhanced migration and invasion of multiple cancers as well as poor patient prognosis. Promising results from in vitro experimental studies using docosahexaenoic acid (DHA) in breast cancer and recombinant porcine NK-lysin A in hepatocellular carcinoma have revealed that anticancer drugs targeting FSCN1 have significant potential clinical applications. This review discusses FSCN1 in terms of five aspects: structure and function, biological processes, regulatory mechanisms, clinical applications, and future prospects.
... Fascin is an actin-bundling protein whose expression is increased during DC maturation (Mosialos et al., 1996;Al-Alwan et al., 2001a;Yamashiro, 2012). In mature DCs, fascin, which can localize to the IS(DC), controls dendrite formation (Al-Alwan et al., 2001a,b;Rothoeft et al., 2006; Figure 1). ...
Book
Full-text available
From a structural point a view, the cytoskeleton has been classically described in relation to its polymerization and depolymerization balance via the availability of monomers/heterodimers and regulators such as; adaptors, chaperones, kinases and phosphatases. However, the dynamic features of the cytoskeletal structures are highly controlled by extracellular cues and intrinsic processes including the cell cycle. Cells with high capacity of regulating their cytoskeleton adapt better to changes in their environment. Thus the dynamics of the cytoskeleton is directly related to the regulation of molecules and organelle polarization and function. Increasing evidence continues to illustrate the relevant role of fine-tuning the cytoskeleton in cell competition, resulting in selection during processes including, for instance, organ development, tumor growth and immune responses. The intracellular organization and adoption of different, polarized shapes constitute the basis for specialized cellular functions, such as migration and directed secretion. Indeed, intracellular compartmentalization allows control of cell metabolism, as observed for mitochondrial contacts with the endoplasmic reticulum and peroxisomes, and the Golgi Apparatus. Regulation of organelle and cell polarization requires specific control of polarized anterograde and retrograde traffic of vesicles. This Research Topic will address physiological and pathological regulation of cytoskeleton dynamics and organelle function during tissue development, tumorigenesis, and immune responses. We welcome submissions of the following article types: Brief Research Report, Hypothesis and Theory, Methods, Mini Review, Opinion, Original Research, Perspective, and Review that fall under the following aspects: • Propagation of plasma membrane signaling through cytoskeleton dynamics • De novo synthesis of cytoskeleton components during cell polarization events • Cytoskeletal dynamics regulating cell division and polarization • Inter-organelle interaction regulated by Cystokeleton dynamics • Regulation of cell polarization by cytoskeleton-organelle contacts
... In adults, FSCN1 is absent or at low levels in normal epithelial cells, and its expression is limited to the neuronal, endothelial, mesenchymal, dendritic, and immune cells [18]. During embryogenesis, FSCN1 is largely expressed in the nervous systems (neuroblasts, melanoblasts, mesenchymal tissue, microcapillary endothelial cells, and antigen-presenting dendritic cells) [18,34,35]. ...
Article
Full-text available
Fascin is an actin-binding protein that is encoded by the FSCN1 gene (located on chromosome 7). It triggers membrane projections and stimulates cell motility in cancer cells. Fascin overexpression has been described in different types of human cancers in which its expression correlated with tumor growth, migration, invasion, and metastasis. Moreover, overexpression of fascin was found in oncovirus-infected cells, such as human papillomaviruses (HPVs) and Epstein-Barr virus (EBV), disrupting the cell–cell adhesion and enhancing cancer progression. Based on these findings, several studies reported fascin as a potential biomarker and a therapeutic target in various cancers. This review provides a brief overview of the FSCN1 role in various cancers with emphasis on gynecological malignancies. We also discuss fascin interactions with other genes and oncoviruses through which it might induce cancer development and progression.
... Fascin is an actin-bundling protein whose expression is increased during DC maturation (Mosialos et al., 1996;Al-Alwan et al., 2001a;Yamashiro, 2012). In mature DCs, fascin, which can localize to the IS(DC), controls dendrite formation (Al-Alwan et al., 2001a,b;Rothoeft et al., 2006; Figure 1). ...
Article
Full-text available
Dendritic cells (DCs) are considered the most potent antigen-presenting cells. DCs control the activation of T cells (TCs) in the lymph nodes. This process involves forming a specialized superstructure at the DC-TC contact zone called the immunological synapse (IS). For the sake of clarity, we call IS(DC) and IS(TC) the DC and TC sides of the IS, respectively. The IS(DC) and IS(TC) seem to organize as multicentric signaling hubs consisting of surface proteins, including adhesion and costimulatory molecules, associated with cytoplasmic components, which comprise cytoskeletal proteins and signaling molecules. Most of the studies on the IS have focused on the IS(TC), and the information on the IS(DC) is still sparse. However, the data available suggest that both IS sides are involved in the control of TC activation. The IS(DC) may govern activities of DCs that confer them the ability to activate the TCs. One key component of the IS(DC) is the actin cytoskeleton. Herein, we discuss experimental data that support the concept that actin polarized at the IS(DC) is essential to maintaining IS stability necessary to induce TC activation.
... Loss of Fascin impairs dendritic cell maturation and therefore motility [39]. During embryonic and fetal development, Fascin is widely expressed in the nervous system, including neuroblasts, neural crest cells, melanoblasts, mesenchymal tissues, microcapillary endothelial cells, gastrointestinal tract, and antigen-presenting dendritic cells [14,[40][41][42][43][44]. A common feature of many of these cell types is that they undergo significant cell migrations, highlighting the importance of Fascin in promoting cell migration. ...
Article
Full-text available
Fascin, an actin-binding protein, regulates many developmental migrations and contributes to cancer metastasis. Specifically, Fascin promotes cell motility, invasion, and adhesion by forming filopodia and invadopodia through its canonical actin bundling function. In addition to bundling actin, Fascin has non-canonical roles in the cell that are thought to promote cell migration. These non-canonical functions include regulating the activity of other actin-binding proteins, binding to and regulating microtubules, mediating mechanotransduction to the nucleus via interaction with the Linker of the Nucleoskeleton and Cytoskeleton (LINC) Complex, and localizing to the nucleus to regulate nuclear actin, the nucleolus, and chromatin modifications. The many functions of Fascin must be coordinately regulated to control cell migration. While much remains to be learned about such mechanisms, Fascin is regulated by post-translational modifications, prostaglandin signaling, protein–protein interactions, and transcriptional means. Here, we review the structure of Fascin, the various functions of Fascin and how they contribute to cell migration, the mechanisms regulating Fascin, and how Fascin contributes to diseases, specifically cancer metastasis.
Article
Full-text available
Jamaican fruit bats (Artibeus jamaicensis) naturally harbor a wide range of viruses of human relevance. These infections are typically mild in bats, suggesting unique features of their immune system. To better understand the immune response to viral infections in bats, we infected male Jamaican fruit bats with the bat-derived influenza A virus (IAV) H18N11. Using comparative single-cell RNA sequencing, we generated single-cell atlases of the Jamaican fruit bat intestine and mesentery. Gene expression profiling showed that H18N11 infection resulted in a moderate induction of interferon-stimulated genes and transcriptional activation of immune cells. H18N11 infection was predominant in various leukocytes, including macrophages, B cells, and NK/T cells. Confirming these findings, human leukocytes, particularly macrophages, were also susceptible to H18N11, highlighting the zoonotic potential of this bat-derived IAV. Our study provides insight into a natural virus-host relationship and thus serves as a fundamental resource for future in-depth characterization of bat immunology.
Article
Rheumatoid arthritis (RA) synovium was identified as “tumor-like” tissues because of the hypoxic microenvironment, significant cell proliferation, and invasion phenotypes. It was reported that hypoxia promoted tumor aggressiveness via up-regulated expression of fascin-1 in cancer. However, the role of fascin-1 in RA synovial hyperplasia and joint injury progression remains unknown. In the current study, we first identified that both fascin-1 and HIF-1α were highly expressed in the RA synovium, in which they were widely colocalized, compared to osteoarthritis(OA). As well, levels of fascin-1 in RA fibroblast-like synoviocytes(FLSs) were found significantly higher than those in OA FLSs. Further, it was demonstrated that the mRNA and protein levels of fascin-1 in RA FLSs were up-regulated in hypoxia (3 % O2) and experimental hypoxia induced by cobalt chloride. Mechanistically, the HIF-1α-mediated hypoxia environment activated the gene expression of the fascin-1 protein, which in turn promoted the migration and invasion of RA FLSs. Accordingly, the restoration of FLSs migration and invasion was observed following siRNA-mediated silencing of fascin-1 and HIF-1α expression. Notably, under the experimental hypoxia, we found that the expression levels of fascin-1, HIF-1α, and p-STAT3 were increased in a time-dependent manner, and fascin-1and HIF-1α expressions were dependent on p-STAT3. Our results indicated that hypoxia-induced fascin-1 up-regulation promoted RA FLSs migration and invasion through the STAT3/HIF-1α/fascin-1 axis, which might represent a novel therapeutic target for the treatment of RA.
Article
Migration of mature dendritic cells (DCs) to lymph nodes is critical for the initiation of adaptive immunity. CCR7, a G-protein-coupled receptor for CCL19/21 chemokines, is known to be essential for chemotaxis of mature DCs, but the molecular mechanism linking inflammation to chemotaxis remains unclear. We previously demonstrated that fascin1, an actin-bundling protein, increases chemotaxis of mature mouse DCs. In this article, we demonstrated that fascin1 enhanced IL-6 secretion and signaling of mature mouse DCs. Furthermore, we demonstrated that IL-6 signaling is required for chemotaxis. Blockage of IL-6 signaling in wild-type DCs with an anti-IL-6 receptor α (IL-6Rα) Ab inhibited chemotaxis toward CCL19. Likewise, knockout of IL-6Rα inhibited chemotaxis of bone marrow-derived DCs. The addition of soluble IL-6Rα and IL-6 rescued chemotaxis of IL-6Rα knockout bone marrow-derived DCs, underscoring the role of IL-6 signaling in chemotaxis. We found that IL-6 signaling is required for internalization of CCR7, the initial step of CCR7 recycling. CCR7 recycling is essential for CCR7-mediated chemotaxis, explaining why IL-6 signaling is required for chemotaxis of mature DCs. Our results have identified IL-6 signaling as a new regulatory pathway for CCR7/CCL19-mediated chemotaxis and suggest that rapid migration of mature DCs to lymph nodes depends on inflammation-associated IL-6 signaling.
Article
Full-text available
The behavior of Langerhans cells (LC) has been examined after skin transplantation and in an organ culture system. Within 24 h (and even within 4 h of culture), LC in epidermal sheets from allografts, isografts, and explants dramatically increased in size and expression of major histocompatibility complex class II molecules, and their numbers were markedly decreased. Using a new procedure, dermal sheets were then examined. By 24 h, cells resembling LC were found close to the epidermal-dermal junction, and by 3 d, they formed cords in dermal lymphatics before leaving the skin. In organ culture, the cells continued to migrate spontaneously into the medium. These observations establish a direct route for migration of LC from the epidermis into the dermis and then out of the skin. These processes are apparently induced by a local inflammatory response, and are independent of host-derived mediators. The phenotype of migratory cells was then examined by two-color immunocytochemistry and FACS analysis. The majority of migratory leukocytes were Ia+ LC, the remainder comprised Thy-1+, CD3+, CD4-, CD8- presumptive T cell receptor gamma/delta+ dendritic epidermal cells, which clustered with the LC, and a small population of adherent Ia-, FcRII+, CD11a/18+ macrophages. In contrast to the cells remaining within the epidermis of grafted skin at 1 d, the migratory cells were heterogeneous in phenotype, particularly with respect to F4/80, FcRII, and interleukin 2 receptor alpha expression, which are useful markers to follow phenotypic maturation of LC. Moreover, cells isolated from the epidermis of grafts at 1 d were more immunostimulatory in the allogeneic mixed leukocyte reaction and oxidative mitogenesis than LC isolated from normal skin, though less potent than spleen cells. The day 1 migratory cells were considerably more immunostimulatory than spleen cells, and day 3-5 migratory cells even more so, suggesting that functional maturation continues in culture. Thus, maturation of LC commences in the epidermis and continues during migration, but the cells do not need to be fully mature in phenotype or function before they leave the skin. In vivo, the migration of epidermal LC via the dermis into lymphatics and then to the draining nodes, where they have been shown previously to home to T areas, would provide a powerful stimulus for graft rejection.
Article
Full-text available
The ability of the human immunodeficiency virus type 1 (HIV-1) to replicate in primary blood dendritic cells was investigated. Dendritic cells compose less than 1% of the circulating leukocytes and are nondividing cells. Highly purified preparations of dendritic cells were obtained using recent advances in cell fractionation. The results of these experiments show that dendritic cells, in contrast to monocytes and T cells, support the active replication of all strains of HIV-1 tested, including T-cell tropic and monocyte/macrophage tropic isolates. The dendritic cell cultures supported much more virus production than did cultures of primary unseparated T cells, CD4+ T cells, and adherent as well as nonadherent monocytes. Replication of HIV-1 in dendritic cells produces no noticeable cytopathic effect nor does it decrease total cell number. The ability of the nonreplicating dendritic cells to support high levels of replication of HIV-1 suggests that this antigen-presenting cell population, which is also capable of supporting clonal T-cell growth, may play a central role in HIV pathogenesis, serving as a source of continued infection of CD4+ T cells and as a reservoir of virus infection.
Article
Full-text available
The behavior of Langerhans cells (LC) has been examined after skin transplantation and in an organ culture system. Within 24 h (and even within 4 h of culture), LC in epidermal sheets from allografts, isografts, and explants dramatically increased in size and expression of major histocompatibility complex class II molecules, and their numbers were markedly decreased. Using a new procedure, dermal sheets were then examined. By 24 h, cells resembling LC were found close to the epidermal-dermal junction, and by 3 d, they formed cords in dermal lymphatics before leaving the skin. In organ culture, the cells continued to migrate spontaneously into the medium. These observations establish a direct route for migration of LC from the epidermis into the dermis and then out of the skin. These processes are apparently induced by a local inflammatory response, and are independent of host-derived mediators. The phenotype of migratory cells was then examined by two-color immunocytochemistry and FACS analysis. The majority of migratory leukocytes were Ia+ LC, the remainder comprised Thy-1+, CD3+, CD4-, CD8- presumptive T cell receptor gamma/delta+ dendritic epidermal cells, which clustered with the LC, and a small population of adherent Ia-, FcRII+, CD11a/18+ macrophages. In contrast to the cells remaining within the epidermis of grafted skin at 1 d, the migratory cells were heterogeneous in phenotype, particularly with respect to F4/80, FcRII, and interleukin 2 receptor alpha expression, which are useful markers to follow phenotypic maturation of LC. Moreover, cells isolated from the epidermis of grafts at 1 d were more immunostimulatory in the allogeneic mixed leukocyte reaction and oxidative mitogenesis than LC isolated from normal skin, though less potent than spleen cells. The day 1 migratory cells were considerably more immunostimulatory than spleen cells, and day 3-5 migratory cells even more so, suggesting that functional maturation continues in culture. Thus, maturation of LC commences in the epidermis and continues during migration, but the cells do not need to be fully mature in phenotype or function before they leave the skin. In vivo, the migration of epidermal LC via the dermis into lymphatics and then to the draining nodes, where they have been shown previously to home to T areas, would provide a powerful stimulus for graft rejection.
Article
Full-text available
In a previous paper from this laboratory, the production of monoclonal antibodies recognizing antigenic determinants common to the alpha and beta chains of bovine brain S-100 protein was reported. In the present study, the immunohistochemical labeling patterns of these monoclonal antibodies against a wide range of normal and pathologic human tissues are described, and these results are compared with those obtained using polyclonal anti-S-100 antiserum. Although many of the reactions of the monoclonal antibodies were very similar to those of the polyclonal antiserum (and the previously reported sites of S-100 immunoreactivity) several additional cell types (e.g., thyroid follicular cells, biliary epithelium, pancreatic cells, renal tubules) were labeled by one or both of the monoclonal antibodies. Blocking experiments prove that immunohistochemical differences obtained with monoclonal and polyclonal antibodies are at least partly caused by differences in the repertoire of antigenic determinants on S-100 protein that are recognized by the two species (i.e., mouse and rabbit, respectively). These results indicate that S-100 may be more widespread in human tissues than previously thought, and that its value as a marker of the histogenetic origin of human tumors should be reappraised. It is suggested that the marked variability observed in the reactivity of different tissues in the present study may indicate that S-100 is a heterogenous group of molecules, and its expression may be related to the functional activity of cells.
Article
A novel human mRNA whose expression is induced over 200-fold in B lymphocytes by latent Epstein-Barr virus (EBV) infection was reverse transcribed, cloned, and sequenced. The mRNA is predicted to encode a protein containing four peptides which precisely match amino acid sequences from a previously identified 55-kDa actin-bundling protein, p55. In vitro translation of the cDNA results in a 55-kDa protein which binds to actin filaments in the presence of purified p55 from HeLa cells. The p55 mRNA is undetectable in non-EBV-infected B- and T-cell lines or in a myelomonocytic cell line (U937). Newly infected primary human B lymphocytes, EBV-transformed B-cell lines, latently infected Burkitt tumor cells expressing EBNA2 and LMP1, a chronic myelogenous leukemia cell line (K562), and an osteosarcoma cell line (TK143) contain high levels of p55 mRNA or protein. In EBV-transformed B cells, p55 localizes to perinuclear cytoplasm and to cell surface processes that resemble filopodia. The p55 mRNA is detected at high levels in spleen and brain tissues, at moderate levels in lung and placenta tissues, and at low levels in skeletal muscle, liver, and tonsil tissues and is undetectable in heart, kidney, pancreas, and bone marrow tissues. Immunohistochemical staining of human brain tissue demonstrates p55 localization to the perinuclear cytoplasm and dendritic processes of many, but not all, types of cortical or cerebellar neurons, to glial cells, and to capillary endothelial cells. In cultured primary rat neurons, p55 is distributed throughout the perinuclear cytoplasm and in subcortical filamentous structures of dendrites and growth cones. p55 is highly evolutionarily conserved since it shows 40% amino acid sequence identity to the Drosophila singed gene product and 37% identity to fascin, an echinoderm actin bundling protein. The evolutionary conservation of p55 and its lack of extensive homology to other actin-binding proteins suggest that p55 has specific microfilament-associated functions in cells in which it is differentially expressed, including neural cells and EBV-transformed B lymphocytes.
Article
Freshly isolated murine epidermal Langerhans cells (LC) are weak stimulators of resting T cells. Upon culture their phenotype changes, their stimulatory activity increases significantly, and they come to resemble lymphoid dendritic cells. Resident murine LC, therefore, might represent a reservoir of immature dendritic cells. We have now used enzyme cytochemistry, a panel of some 80 monoclonal antibodies, and immunofluorescence microscopy or two-color flow cytometry, as well as transmission electron microscopy, to analyse the phenotype and morphology of human LC before and after 2–4 d of bulk epidermal cell culture. In addition, LC were enriched from bulk epidermal cell cultures, and their stimulatory capacity was tested in the allogeneic mixed leukocyte reaction and the oxidative mitogenesis assay. Cultured human LC resembled human lymphoid dendritic cells in morphology, phenotype, and function. Specifically, LC became non-adherent upon culture and developed sheet-like processes (so-called "veils"), decreased their surface ATP/ ADP'ase activity, and lost nonspecific esterase activity. As in the mouse, surface expression of MHC class I and II antigens increased significantly, and FcII receptors were significantly reduced. Markers that are expressed by dendritic cells (like CD40) appeared on LC following culture. Cultured human LC were potent T-cell stimulators. Our findings support the view that resident human LC, like murine LC, represent immature precursors of lymphoid dendritic cells in skin-draining lymph nodes.
Article
cDNA isolated from a human lymphocyte library were analyzed and shown to encode a novel cell-surface glycoprotein, termed HB15, expressed by dendritic cell subsets and activated lymphocytes. The predicted mature 186 amino acid protein was composed of a single extracellular V-type Ig-like domain, a transmembrane region, and a 39-amino acid cytoplasmic domain. In contrast to most Ig-like domains, analysis of a partial genomic DNA clone revealed that the extracellular Ig-like domain of HB15 was encoded by at least two exons. Northern blot analysis revealed that HB15 derived from three mRNA transcripts of approximately 1.7, 2.0, and 2.5 kb expressed by lymphoblastoid cell lines. Two mAb reactive with HB15 were produced and used to show that HB15 is expressed as a single chain cell-surface glycoprotein of M(r) 45,000. HB15 expression was specific for lymphoblastoid cell lines and mitogen-activated lymphocytes, and HB15 was not expressed at detectable levels by circulating leukocytes. Immunohistologic analysis revealed that HB15 had a unique pattern of expression, being found predominantly in hemopoietic tissues with strong expression by scattered interfollicular interdigitating reticulum cells and weak expression by germinal center cells. HB15 was also expressed by Langerhans cells within the skin. HB15 therefore serves as a unique marker for the subset of dendritic cells represented by Langerhans cells and interdigitating reticulum cells. Thus, the HB15 glycoprotein represents a newly identified member of the Ig superfamily that may play a significant role in Ag presentation or the cellular interactions that follow lymphocyte activation.
Article
This chapter discusses the role of follicular dendritic cells in lymphoid tissues. Characteristic features of primary and secondary lymphoid follicles are follicular dendritic cells (FDCs) that are not found in any other human organ. FDCs were first described as a nonlymphoid population of embryonic nonphagocytic reticulum cells. Other names for FDCs have been used in the past, for example, dendritic reticulum cells (DRCs), follicular dendritic reticulum cells, antigen-retaining reticular cells, follicular antigen-binding dendritic cells, and dendritic macrophages. FDCs are considered as having a passive role in the function of the germinal center. It is characterized as nonphagocytic cells that capture and retain complexes of antigen, antibodies, and C3 on their cell surface. By expressing antigen–antibody complexes and a large number of cell–cell and cellmatrix molecules, FDCs were shown to regulate normal B cell function. There is growing evidence that FDCs could be important constituents of the malignant counterpart of the germinal center. In addition, FDCs were demonstrated to be the target cells in human immunodeficiency virus (HIV)-related lymphadenopathy. These data can be integrated into a concept of FDCs as the key component of the germinal center microenvironment. FDCs demonstrate a unique antigen pattern by co-expressing myeloid and B cell markers.
Article
Dendritic cells are a system of antigen presenting cells that function to initiate several immune responses such as the sensitization of MHC-restricted T cells, the rejection of organ transplants, and the formation of T-dependent antibodies. Dendritic cells are found in many nonlymphoid tissues but can migrate via the afferent lymph or the blood stream to the T-dependent areas of lymphoid organs. In skin, the immunostimulatory function of dendritic cells is enhanced by cytokines, especially GM-CSF. After foreign proteins are administered in situ, dendritic cells are a principal reservoir of immunogen. In vitro studies indicate that dendritic cells only process proteins for a short period of time, when the rate of synthesis of MHC products and content of acidic endocytic vesicles are high. Antigen processing is selectively dampened after a day in culture, but the capacity to stimulate responses to surface bound peptides and mitogens remains strong. Dendritic cells are motile, and efficiently cluster and activate T cells that are specific for stimuli on the cell surface. High levels of MHC class-I and -II products and several adhesins, such as ICAM-1 and LFA-3, likely contribute to these functions. Therefore dendritic cells are specialized to mediate several physiologic components of immunogenicity such as the acquisition of antigens in tissues, the migration to lymphoid organs, and the identification and activation of antigen-specific T cells. The function of these presenting cells in immunologic tolerance is just beginning to be studied.
Article
Epstein-Barr virus (EBV) has been associated with serious or fatal lymphoproliferative disease in immunocompromised patients. EBV nuclear protein 2 and latent membrane protein are characteristically expressed in B lymphocytes proliferating in vitro in response to growth transformation by EBV. These two proteins are thought to be effectors of lymphocyte growth since they increase the expression of B-lymphocyte activation (CD23) and cell-adhesion (LFA 3 and ICAM 1) molecules in vitro. Using monoclonal antibody-immune microscopy, we have demonstrated that these two EBV proteins and their associated B-lymphocyte activation or adhesion molecules are expressed in the infiltrating B lymphocytes in immunocompromised patients with EBV lymphoproliferative disease. These monoclonal antibodies should be useful in the early diagnosis of EBV lymphoproliferative disease and in distinguishing it from other B-lymphocyte cancers associated with EBV, such as Burkitt's lymphoma. The finding of EBV nuclear protein 2 and latent membrane protein and their associated activation or adhesion molecules provides a further pathophysiologic link between EBV and the proliferation of B lymphocytes in immunocompromised patients.