ArticlePDF Available

Partial Mediation of Glucocorticoid Antiproliferate Effects by Lipocortins

Authors:

Abstract

The glucocorticoids (GCs) dexamethasone (DEX) and prednisolone (PRED), in a concentration-dependent fashion, profoundly inhibit mitogen-induced proliferation of human peripheral blood mononuclear lymphocytes (PBML). This inhibition was specific for GCs, as non-GC steroids were devoid of any antiproliferative capacity. GCs enhanced the mRNA (Northern blot) and protein (Western blot) expression of the calcium and phospholipid binding proteins lipocortin I, II, and V. As a consequence of mitogenic stimulation, PBML secrete PGE2 and leukotriene B4 (LTB4). Antiproliferative concentrations of both DEX and PRED as well as recombinant lipocortin I abolished PGE2 and LTB4 production, suggesting an involvement of lipocortins in GC-mediated antiproliferative effects, possibly by inhibiting eicosanoid production and, consequently, mitogen-induced cellular proliferation. Whereas 5,8,11,14-eicosatetraynoic acid and nordihydroguaiaretic acid mimicked DEX and PRED in inhibiting PGE2 and LTB4 production, neither 5,8,11,14-eicosatetraynoic acid nor nordihydroguaiaretic acid had any effect on mitogen-induced PBML proliferation, indicating that the GC-mediated antiproliferative effect is separate from their effects on eicosanoid release. Furthermore, neutralizing anti-lipocortin I and anti-lipocortin II mAb, while reversing the inhibitory activity of DEX and PRED on PGE2 and LTB4 production, only partially reversed DEX- and PRED-mediated antiproliferative effects. This indicates that the GC-mediated antiproliferative effect is not dependent on inhibition of eicosanoid release by lipocortins and suggests the existence of lipocortin-dependent and lipocortin-independent pathways by which GCs mediate their antiproliferative effects.
of March 10, 2010
This information is current as
1996;157;5231-5239 J. Immunol.
Barth and TB Strom
WY Almawi, MS Saouda, AC Stevens, ML Lipman, CM
antiproliferative effects by lipocortins
Partial mediation of glucocorticoid
References
http://www.jimmunol.org#otherarticles
2 online articles that cite this article can be accessed at:
Subscriptions
http://www.jimmunol.org/subscriptions/online at
isThe Journal of ImmunologyInformation about subscribing to
Permissions
http://www.aai.org/ji/copyright.html
Submit copyright permission requests at
Email Alerts
http://www.jimmunol.org/subscriptions/etoc.shtmlup at
Receive free email alerts when new articles cite this article. Sign
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Immunologists, Inc. All rights reserved.
Copyright ©1996 by The American Association of
Rockville Pike, Bethesda, MD 20814-3994.
The American Association of Immunologists, Inc., 9650
is published twice each month byThe Journal of Immunology
on March 10, 2010 www.jimmunol.orgDownloaded from
Partial Mediation of Glucocorticoid Antiproliferative Effects
by
Lipocortins'
Wassim
Y.
AImawi,** Myrna
S.
Saouda,* Anthony C. Stevens,t Mark
1.
Lipman,+
Claudia
M.
Barth,t and Terry
B.
Stromt
The glucocorticoids (GCs) dexamethasone
(DEX)
and prednisolone (PRED), in a concentration-dependent fashion, profoundly
inhibit mitogen-induced proliferation of human peripheral blood mononuclear lymphocytes (PBML). This inhibition was specific
for GCs, as non-GC steroids were devoid of any antiproliferative capacity. GCs enhanced the mRNA (Northern blot) and protein
(Western blot) expression of
the
calcium and phospholipid binding proteins lipocortin
I,
II,
and
V.
As a consequence of
mitogenic stimulation, PBML secrete PGE, and leukotriene B, (LTB,). Antiproliferative concentrations of both
DEX
and PRED
as well as recombinant lipocortin
I
abolished PGE, and
LTB,
production, suggesting an involvement of lipocortins in GC-
mediated antiproliferative effects, possibly by inhibiting eicosanoid production and, consequently, mitogen-induced cellular
proliferation. Whereas
5,8,11,14-eicosatetraynoic acid and nordihydroguaiaretic acid mimicked
DEX
and
PRED
in inhibiting
PGE,
and LTB, production, neither 5,8,11,14-eicosatetraynoic acid nor nordihydroguaiaretic acid had any effect on mitogen-
induced PBML proliferation, indicating that the GC-mediated antiproliferative effect
is
separate from their effects on eicosanoid
release. Furthermore, neutralizing anti-lipocortin
I
and anti-lipocortin
II
mAb, while reversing the inhibitory activity of
DEX
and
PRED on PGE, and LTB, production, only partially reversed DEX- and PRED-mediated antiproliferative effects. This indicates
that the GC-mediated antiproliferative effect
is
not dependent on inhibition of eicosanoid release by lipocortins and suggests the
existence of lipocortin-dependent and lipocortin-independent pathways by which GCs mediate their antiproliferative effects.
The
Journal
of Immunology, 1996, 157: 5231-5239.
G
lucocorticoids (GCS),~ as anti-inflammatory and immu-
nosuppressive agents, are used in treating autoimmune
diseases and graft rejection episodes
(1,
2). However,
despite their wide-spread use, the precise mechanism by which
GCs
mediate their antiproliferative effects remains evasive due in
part to the myriad of biological effects mediated by the GCs.
Among the mechanisms postulated for GC-mediated antiprolifera-
tive effects include blockade of activation-associated increases in
transmembrane ionic fluxes
(3,
4),
alteration in membrane lipid
phospholipid profile (5,
6),
inhibition of cytokine gene expression
(7-9),
andor induction of the calcium and phospholipid binding
proteins, the lipocortins
(10,
11).
It is well documented that GCs induce lipocortin expression at
the mRNA and protein levels. Lipocortins, due to their capacity to
inhibit phospholipase
A,
(PLA,) activity
(12,
13),
block arachi-
donic acid (AA) release from membrane-bound stores, resulting in
*Department of Biochemistry, Faculty
of
Medicine, American University of
Medicine, Beth Israel Hospital and Harvard Medical School, Boston, MA 02215
Beirut, Beirut, Lebanon; and 'Division
of
Clinical Immunology, Department of
ber 5, 1996.
Received for publication November 30, 1995. Accepted for publication Septem-
The costs of publication of this article were defrayed in part by the payment of
page charges. This article
must
therefore be hereby marked advertisement in
accordance with 18 U.S.C. Section
1
734 solely
to
indicate this fact.
'
This work
was
supported by grants from the National Institutes of Health, the
Kidney Foundation
of
Canada, and the American University
of
Beirut-M.P.P.
partment
of
Biochemistry, Faculty
of
Medicine, American University of Beirut,
Address correspondence and reprlnt requests
to
Dr. Wassim
Y.
Almawi, De-
850 Third Ave., New York, NY 10022-6222,
Abbreviations used in this paper: GCs, glucocorticoids; PLA,, phospholipase
A,; LTB,, leukotriene B,;
DEX,
dexamethasone; PRED, prednisolone; PBML, pe-
rlpheral blood mononuclear lymphocytes; ETYA, eicosatetraynoic acid; NDGA,
nordihydroguaiaretic acid; AA, arachldonic acid.
Copyright
0
1996
by
The American Association
of
Immunologists
the inhibition of PC and leukotriene (LT) production
(14,
15). This
prompted the conclusion that GCs exert their effects through
li-
pocortin induction, which, in turn, blocks
AA
release and, conse-
quently,
PC
and LT production
(IO,
121, resulting in the suppres-
sion of select elements of the signal transduction pathway(s)
(I
3).
Other reports challenged this conclusion by presenting data show-
ing that GC-mediated suppression may be a separate event from
classical lipocortin induction, assessed by the inhibition of eico-
sanoid production (15-17). This did not rule out a possible in-
volvement of lipocortins in transducing GC-mediated effects via
an, as yet, undisclosed mechanism
(16,
18).
Previously, we demonstrated that GC-mediated antiproliferative ef-
fects do not involve altering the generation
of
second messenger sys-
tems (rise in intracellular calcium, activation and translocation of
protein kinase C from cytosolic to membrane-bound compart-
ments) that operate as a consequence of cellular activation
(19).
GCs mediate their antiproliferative effects by inhibiting cytokine
expression
as
1) antiproliferative concentrations of dexamethasone
(DEX) and prednisolone (PRED) blocked steady state IL-I
(9),
IL-2
(7),
and IFN-y
(8)
mRNA expression; and 2) the combination
of IL-1,
IL-6,
and IFN-y completely abrogated GC-mediated an-
tiproliferative effects (7). Here we show that lipocortins mediate in
part GC-mediated antiproliferative effects, indicating the existence
of a lipocortin-dependent and independent pathways by which
GCs mediate their effects.
Materials and Methods
Preparation
of
PBML
Venous blood from healthy volunteers was diluted 1/2 in saline, layered on
Hypaque-Ficoll
(S.G.
1.077, Pharmacia Fine Chemicals, Piscataway,
NJ),
and centrifuged for 20 min at 2000
X
g. The
interphase containing
PBML
was washed three times in saline and was resuspended at
IO6
cells/ml
in
0022-1 767/96/$02.00
on March 10, 2010 www.jimmunol.orgDownloaded from
5232
PARTIAL MEDIATION
OF
GLUCOCORTICOID EFFECTS
BY
LIPOCORTINS
RPMI 1640 culture medium (M. A. Bioproducts, Bethesda, MD)
supple-
mented with 50 pM @ME (Sigma Chemical Co.,
St.
Louis, MO), 2 mM
L-glutamine (Life Technologies, Grand Island, NY), penicillin-streptomy-
cin (Life Technologies) at 100 IU/ml and
100
pg/ml, respectively, and
10%
(v/v)
human type AB serum (M. A. Bioproducts), referred to hereafter
as
complete medium.
Reagents and Abs
PMA and leupeptin were purchased from Sigma Chemical Co., and PHA
was obtained from Difco Laboratories (Detroit, MI). Stock solutions of
DEX, PRED, AA,
5,8,11,14-eicosatetraynoic
acid (ETYA), nordihydrogui-
aretic acid (NDGA), cholesterol, /3-estradiol, and aldosterone were made in
70% ethanol and stored at -20°C until used (all obtained from Sigma
Chemical Co.). All other steroids used in the study were supplied as oil-
based injections from the Beth Israel Hospital Pharmacy (Boston, MA).
Anti-lipoconin
I
(lA),
11,
and
V
mAb were obtained from Biogen, Inc.
(Cambridge, MA), courtesy of Dr.
J.
Browning. Gold-conjugated goat anti-
mouse Abs were obtained from Bio-Rad (Mississauga, Ontario, Canada),
and mouse IgGl was purchased from Ortho Pharmaceutical Corp.
(Raritan, NJ).
Proliferation assays
For
mitogen-induced proliferation PBML (5
X
IO5
cells/ml) were cultured
in 96-well, flat-bottom microtiter plates (Nunc, Burlington, Ontario, Can-
ada) and were stimulated with PHA (5 pg/ml) and PMA (5 ng/ml) or with
Con A
(10
pg/ml).
The
cells were incubated for 72 h at 37°C in a
5%
CO,
humidified atmosphere.
For
MLR, PBML
(lo6
cells/ml) were cocultured
with mitomycin C-treated (25 pg/ml; Sigma Chemical
Co.)
allogeneic cells
(lo6 celldml) in complete medium containing 50 pM
@ME
(Sigma Chem-
ical Co.) for 5 days at 37°C. [3H]TdR (1 pCi/well; New England Nuclear,
Boston, MA) was added during the last 4 h of the culture period, and
proliferation was determined by measuring the cellular uptake of [3H]TdR
by liquid scintillation.
Determination of
LTB,
and
PGE,
PBMC were cultured in 24-well, flat-bottom plates (Falcon, Lincoln Park,
NJ) at 5
X
lo5 cells/ml in complete medium.
The
cells were pretreated with
the indicated agents for 4 h at 37T, followed by stimulation with PHA plus
PMA. Cellfree supernatants of cultured and treated PBML were assayed
for LTB, and
PGE
by RIA using a commercially available kit from Am-
ersham Cop (Arlington Heights,
IL).
All determinations were performed
in duplicate.
Cellular fractionation
PBML
(lo6
cellslml) were stimulated with PHA (5 pg/ml) and PMA (5
ng/ml), cultured for 24 h at 37°C with
or
without test drugs, washed
twice in HBSS (Life Technologies, Grand Island, NY), and resuspended
at lo7 cells/ml in extraction buffer (20 mM Tris-C1, pH 7.2;
2
mM
EDTA; 50 mM 2-ME; and 100 pg/ml leupeptin). The cells were then
sonicated for 20
s
and centrifuged at
50,000
X
g
for
1
h at 4°C. Cy-
tosolic lipocortin-containing fractions were lyophilized at stored at
-20°C until assayed.
Western blot analysis
Cytosolic lipocortin-containing fractions were subjected to 7.5% SDS-
PAGE according
to
the method of Laemmli (20). Proteins were transferred
to nitrocellulose membranes, and the membranes were incubated with the
primary Ab for 2 h at room temperature. Gold-conjugated goat anti-mouse
mAb was added to washed membranes, and the membranes were incubated
for
18
to 20 h at room temperature.
Northern blot analysis
Total cellular RNA was extracted by the guanidium isothiocyanateLiC1
method under strict RNase-free conditions (21).
RNA
(10 pgfiane) was
electrophoresed on a 1% agarose gel containing 2.2 M formaldehyde (22)
and electrotransferred onto Hybond N+ membranes (Amersham). The
membranes were prehybridized at 42°C for
4
h in a prehybridization
so-
lution containing 50%
(v/v)
deionized formamide,
l
X
Denhardt's solution,
I%
SDS,
1
mM NaCI,
5
mM
Tris-CI (pH 7.4), and
10%
dextran sulfate
(Sigma Chemical Co.). [32P]dCTP-labeled (New England Nuclear) cDNA
probes were then added, and the membranes were hybridized at 42°C for
12 to 18 h. After washing twice in
2X
SSC/O.l%
SDS
at 42°C and twice
Concentration
(M)
FIGURE
1.
Specificity
of
the GC-mediated antiproliferative effect.
The proliferation of PBML stimulated with PHA
(5
pghnl)
and PMA
(5
ng/ml) and treated with culture medium (positive control), ethanol (ve-
hicle control), or the indicated steroid at the indicated concentration.
Proliferation was determined
72
h post-culture initiation; control
values were: background cpm,
781
t
124;
positive control cpm,
92,715
-+
9,877;
and ethanol control cpm,
86,552
2
9,140.
Data
points indicate the percent suppression, calculated
as:(l
-
[(test
cpm
-
background cpm)/(control cpm
-
background cpm)l)
x
100%
in
0.1
X
SSC/O.l% SDS at 75°C for
20
min, and the membranes were
exposed to Kodak x-ray film (Eastman Kodak, Rochester, NY) at -70°C
for 24 to 72 h.
Results
Comparative effects
of
GC
and non-GC steroids on mitogen-
induced
T
cell proliferation
The effect of GC and non-GC steroids on mitogen-induced PBML
proliferation was assessed by adding the steroids (and ethanol), at
lo-'
to 10"o M, to PBML cultures stimulated with PHA
(5
fig/
ml) and PMA
(5
ng/ml; referred to thereafter as PHA-PMA) at
culture initiation; proliferation was determined by measuring the
cellular uptake
of
[3H]TdR
72
h postactivation. The results pre-
sented in Figure
1
show that, of all the steroids tested, only DEX
(EC,,
=
5
X
lo-'
M), betamethasone (EC,,
=
7.5
X
lo-'
M),
hydrocortisone (EC,,
=
5
X
10"
M), and PRED (EC,,
=
5
X
M) inhibited mitogen-induced cellular proliferation (EC,,
=
drug concentration yielding
50%
suppression
of
PHA-PMA re-
sponses). In contrast, the non-GC steroids aldosterone, andros-
terone, cholesterol, diethylstilbesterol, /3-estradiol, nandrolone,
pregnenolone, progesterone, and testosterone were devoid of an-
tiproliferative capacity at all concentrations tested, as the prolifer-
ative responses of mitogen-stimulated PBML cultures treated with
non-GC steroids were not statistically different from those
of
either
positive or ethanol control cultures (data not shown).
Suppression
of
mitogen- and alloantigen-induced
T
cell
proliferation by GCs
The antiproliferative capacity of GCs was further investigated by
adding ethanol, DEX, or PRED, at
10"'
to
IO"
M, to PBML
cultures stimulated with Con
A
(10 pg/ml; Fig.
2.4)
or with mit-
omycin C-treated allogeneic cells (MLR; Fig.
2B);
cellular prolif-
eration was determined
3
days
(Con A) or
5
days (MLR) post-
culture initiation. Both DEX and PRED, in a concentration-
dependent fashion, inhibited Con A-induced and MLR-driven T
on March 10, 2010 www.jimmunol.orgDownloaded from
The
Journal
of
Immunology
5233
FIGURE
2.
inhibition by
GCs
of mitogen-
and alloantigen-induced proliferation.
A,
PBML
(5
X
1
O4
cells) were stimulated with
Con
A
(1
o
pg/mI);
6,
PBML
(I
.5
x
1
o5
cells)
were stimulated with mitomycin C-treated
allogeneic cells
(1.5
x
lo5
cells). Both
groups of cells were treated with ethanol
(vehicle control),
DEX,
or PRED. Prolifera-
tion was assessed
3
days
(A)
or
5
days
(B)
postculture initiation. Proliferation values
for
Con
A:
background cpm,
1,123
5
258;
positive control cpm,
108,849
?
11,624;
for
MLR:
background cpm,
51 3
2
126;
positive
control cpm,
74,083
rt
6,466.
Data points
are the mean of eight individually performed
experiments and indicate the percent sup-
pression calculated as follows:
(1
-
[(test
cpm
-
background cpm)/(control cpm
-
background cpm)])
X
100%.
80
-
60
-
40
-
20
-
DEX’
MP’
Concentration
(M)
DEX
MP
..
1
10”O
I
o-~
1
o-8
10”
1
o-6
1
o-~
cell proliferation (Fig.
2,
A
and
B).
Furthermore, DEX was
50-
to
100-fold more potent than PRED in inhibiting both T cell prolif-
erative responses, assessed by comparing
the
EC,, values for both
agents.
GCs
induce lipocortin expression
The effect of GCs on lipocortin
I,
11,
and
V
expression was first
assessed by examining the effect of DEX and PRED on lipocortin
I
and
I1
steady state mRNA expression. Total cellular RNA, ex-
tracted from DEX-treated and mitogen-stimulated cultures, was
subjected to Northern blot analysis using 32P-labeled lipocortin
I
and lipocortin
I1
cDNA probes.
PBML stimulation with PHA-PMA resulted in a reproducible
induction
of
lipocortin
I,
but not lipocortin
11,
mRNA expression.
DEX, in a concentration-dependent fashion, up-regulated lipocor-
tin
I
and lipocortin
I1
mRNA expression, with the maximal re-
Concentration
(M)
sponse seen at to
10”
M (Fig.
3).
Similarly, lipocortin
I
and
I1
mRNA levels were up-regulated in PRED-treated and mitogen-
stimulated PBML cultures (data not shown).
We next assessed whether GC-induced up-regulation in lipocor-
tin
I
and lipocortin
I1
steady state mRNA levels resulted in parallel
increases in lipocortin
I,
lipocortin
11,
and lipocortin
V
protein
secretion. Cytosolic fractions of GC-treated and mitogen-stimu-
lated PBML cultures were subjected to SDS-PAGEmestern blot
analysis, using specific anti-lipocortin
I, 11,
and
V
mAbs. Parallel
to its effect on lipocortin
I
mRNA, stimulation of PBML with
PHA-PMA resulted in the induction of lipocortin
I,
but not lipocor-
tin
I1
or lipocortin
V,
protein secretion (Fig.
4,
A
and
B).
DEX
(Fig.
4A)
and PRED (Fig.
4B)
in a concentration-dependent fash-
ion, enhanced the cytosolic accumulation of lipocortin
I,
li-
pocortin
11,
and lipocortin
V
proteins. Smaller amounts of li-
pocortin
I
and lipocortin
I1
were also detected in the membrane
on March 10, 2010 www.jimmunol.orgDownloaded from
5234
28s
-
18S4
LC
I
-
LC
II-
PHA
PARTIAL MEDIATION
OF
GLUCOCORTICOID EFFECTS
BY
LIPOCORTINS
togen-stimulated PBML cultures were treated with the inhibitors
of AA release, ETYA and NDGA, and their supernatants were
assayed for PGE, and LTB,. NDGA and ETYA,
in
a concentra-
tion-dependent manner, inhibited PGE, (Fig. 7A) and LTB, (Fig.
7B) production
in
a manner analogous to DEX, PRED, and
li-
pocortin
I.
In
contrast to DEX, which inhibited mitogen-induced
proliferation at concentrations as low as M, NDGA and
ETYA, at all concentrations tested, failed to inhibit mitogen-in-
duced T cell proliferation (Fig.
8).
To rule out the possibility that the GC-mediated antiproliferative
effect was due to inhibition of AA release, DEX-treated and mi-
togen-stimulated PBML cultures were reconstituted with exoge-
nous AA, and proliferation was determined 72
h
postincubation.
Fw
The results presented
in
Figure 9 demonstrate that, at all concen-
trations tested, AA did not alter DEX-mediated antiproliferative
0
effects (Fig. 9).
Effect of anti-lipocortin /-neutralizing mAb
on
DEX-induced
0
inhibition of eicosanoid production and cellular proliferation
‘0
To determine whether DEX-induced inhibition of eicosanoid pro-
duction and cellular proliferation was mediated via lipocortins, the
effects of neutralizing anti-lipocortin I and anti-lipocortin I1 mAb
on DEX-mediated blockade of PGE, and LTB, production and on
DEX-induced inhibition of mitogen-induced cellular proliferation
vidually and
in
combination, totally abrogated DEX-induced
inhi-
-
+++
++
were assessed. Anti-lipocortin
I
and anti-lipocortin I1 mAb, indi-
DEX
(M)
-
-
10-5 10-6
10-7
10-8
FIGURE
3.
Northern blot analysis
of
total cellular
RNA.
Total cellu-
lar
RNA
(10
&lane) was extracted from
PBML
stimulated with
PHA-
PMA
and treated with
DEX
at
to
lo-’
M.
Top,
Ethidium
bromide
staining
of
total
RNA
electrophoresed on
a
1%
agarose denaturing
gel.
Following Northern transfer, the membranes were probed with 32P-
labeled lipocortin
I
(middle)
and
lipocortin
II
(bottom)
cDNA and ex-
posed to Kodak x-ray
film
for
24
h
at
-70°C.
fractions of GC-treated and mitogen-stimulated cultures (data
not shown).
Inhibition of eicosanoid production and T cell proliferation
by lipocortin
I
and
GCs
We next investigated whether induction
of
lipocortins by PRED
and DEX results
in
inhibition of AA release and, subsequently,
blockade of mitogen-induced cellular proliferation. Mitogen-stim-
ulated cultures were treated with DEX, PRED, and lipocortin I,
and their supernatants were assayed for PGE, and LTB, by RIA.
DEX, PRED, and lipocortin I,
in
a concentration-dependent fash-
ion, inhibited PGE, (Fig.
5A)
and LTB, (Fig.
5B)
production. The
addition of
50
to
100
pM AA reversed DEX-, PRED-, and
li-
pocortin I-induced blockade
of
PGE, production, indicating that
all three agents inhibit PGE, (and LTB,) production by blocking
the release of AA (data not shown).
We then assessed the effect of lipocortin I on mitogen-induced
cellular proliferation by adding lipocortin I and DEX, at to
M, to mitogen-stimulated cultures. Lipocortin I,
in
a concen-
tration-dependent fashion, inhibited mitogen-induced T cell pro-
liferation (Fig. 6). By comparison to lipocortin I, DEX was
>15-
fold more potent
in
inhibiting T cell proliferation, assessed by
comparing the EC,, values for both agents.
Effects of NDGA and ETYA
on
cellular proliferation and
eicosanoid production
To
assess whether GC-induced antiproliferative effects was due
to
blockade of AA release (resulting from lipocortin induction), mi-
bition of PGE, and LTB, production by mitogen-stimulated
PBML cultures (Fig.
10).
The addition of anti-lipocortin I mAb
or
anti-lipocortin I1 mAb
to DEX-treated and PHA-PMA-stimulated cultures resulted
in
par-
tial abrogation
of
DEX-induced inhibition of PBML proliferation,
which did not exceed 45% of the control values (Fig.
11).
Fur-
thermore, the addition of both anti-lipocortin
I
and anti-lipocortin
I1 mAb did not result
in
any additive or synergistic effect, hence
demonstrating that the GC-mediated antiproliferative effect is par-
tially mediated by lipocortins. Taken together, these results sug-
gest that GC-mediated antiproliferative effects follow lipocortin-
dependent and lipocortin-independent pathways via a mechanism
distinct from the inhibition of AA release.
Discussion
The GCs DEX and PRED, in a concentration-dependent fashion,
profoundly inhibit the proliferation
of
human PBML cultures
in-
duced by mitogenic
or
allogeneic stimuli, and up-regulate lipocor-
tin
mRNA and protein expression. In view of the inducibility of
lipocortins by GCs and the mimicry of GC effects by lipocortin
I,
we investigated whether GC-mediated antiproliferative effects are
mediated by lipocortins.
Insofar as
GCs
are known to suppress mitogen and Ag-induced
cellular proliferation, we tested the capacity of non-GC steroids to
inhibit mitogen-induced cellular proliferation. Immunosuppression
was specific for the GCs, as non-GC steroids did not affect mito-
gen-elicited
or
OKT3-stimulated (7, 19) cellular proliferation. This
is
in
contrast to published reports claiming that P-estradiol (23,
24), cholesterol (25), diethylstilbestrol (26), progesterone (27,28),
testosterone (29). and other non-GC steroids
(30,
31) are immu-
nosuppressive. It should be noted that
in
the studies quoted, ste-
roid-mediated immunosuppression was associated with conditions
such as arthritis (23), cancer (26), pregnancy (27, 28), and adult
thymectomy (29). predisposing factors that may have contributed
to the decreased immunity reported. In contrast and similar to
our
finding, non-GC immunosuppression was either lacking (16,32) or
seen only at toxic concentrations (33) in normal human subjects.
on March 10, 2010 www.jimmunol.orgDownloaded from
The
Journal
of
Immunology
5235
*&
F?
IRRELEVENT
3'
ANTIBODY
I
!
LIPOCORTIN
I
B
PHA+PMA
-
+
+
+
+
PREDNISOLONE
-
-
+
+
+
CONCENTRATION
(M)
10-7
10-6
10-5
Control Antibody
LIPOCORTIN
I
LIPOCORTIN
I1
LIPOCORTIN
V
FIGURE
4.
Western blot analysis. Western blot analysis
of
cytoplasmic preparations
of
unstimulated
PBML
(UNSTIMULATED),
of
PBML
stim-
ulated with
PHA-PMA
(PHNPMA), and
of
PBML
stimulated with
PHA-PMA
and treated with DEX
(A)
or
with PRED
(B)
at
the
indicated con-
centrations. Membranes were hybridized with irrelevant
lgGl
or
with anti-lipocortin I, It, and
V
Abs, and protein-antibody interactions were
visualized by gold staining.
PHA-PMA stimulation was associated with a reproducible in-
togen-stimulated cultures. DEX and PRED, in a concentration-
duction of lipocortin
I,
but not lipocortin
11,
mRNA expression.
dependent fashion, up-regulated lipocortin
I
and induced lipocortin
Insofar as
GCs
up-regulate lipocortin mRNA expression at the
I1
steady state mRNA expression, with maximal effects seen at
transcriptional and post-transcriptional levels
(13,34-38),
here we
IO-"
M. The decline in mRNA expression seen at concentrations
demonstrate that antiproliferative concentrations of DEX and
higher than
IO"
M
is
most likely due to mRNA degradation, as
PRED up-regulate lipocortin mRNA and protein expression in mi-
previously suggested
(IO,
39).
on March 10, 2010 www.jimmunol.orgDownloaded from
5236
PARTIAL MEDIATION
OF
GLUCOCORTICOID EFFECTS
BY
LIPOCORTINS
PGE
Production
c
C
Q,
-1 1
-10
-9
-8
-7
-6
-5
log
Concentration (M)
LTB
Production
,!':iB,
60
-
40
Lr,
Q,
C
L
v
a"
20
0
-1
1
-1
0
-9
-8 -7
-6
-5
log
Concentration
(M)
FIGURE
5.
Inhibition of eicosanoid production by DEX, PRED, and
lipocortin
I.
PGE,
(A) and
LTB,
(5)
levels were determined by
RIA
in
the culture supernatants of
PBML
stimulated with
PHA-PMA
and
treated with
DEX
(circles), lipocortin
1
(squares), and
PRED
(triangles).
Data points represent the mean of five individually performed exper-
iments,
and
indicate suppression, calculated as:
(1
-
[(test conc.
-
background conc.)/(control conc.
-
background conc.)])
X
100%.
DEX and PRED also enhanced the cytosolic accumulation
of
lipocortin
I,
lipococortin
11,
and lipocortin
V
proteins. Similar to
earlier reports, lipocortins
I
and I1 migrated on SDS-PAGE as two
bands with apparent molecular masses of 37 and 33 kDa, respec-
tively (36,40). In our hands, a third band migrating at 31 kDa was
consistently observed only in DEX-treated cultures; the nature and
significance of this band have yet to be determined.
Lipocortin
I
shares with DEX and PRED the capacity to block
PGE, and LTB, synthesis and to inhibit mitogen-induced cellular
proliferation. In view of the inducibility of lipocortins by GCs (34,
36), and the effect of lipocortins as PLA, inhibitors on blocking
AA release and subsequently PG and LT synthesis (13, 41), we
tested whether the GC-mediated antiproliferative effect is due to
the induction of lipocortin expression, which, in turn, is associated
with inhibition of AA release and metabolism. While the PG and
LT inhibitors NDGA and ETYA (42, 43) share with GCs and
lipocortin
I
the capacity to inhibit PGE, and LTB, production,
both NDGA and ETYA failed to inhibit mitogen-induced cellular
proliferation, indicating that GC- and lipocortin 1-mediated
antiproliferative effects were not the result of inhibition of AA
release and metabolism.
Lipocortin I was reported to mediate several GC effects, includ-
ing superoxide generation by A23 187-stimulated macrophages
-
C
Q,
100
-
80
-
60
-
40
-
20
-
DEX
+
LC1
0
,:.1'1
'
"',';-l'o'
'",'b-o
'
'
,';-E
'
"',
b-7
1
b-6
70-5
Concentration
(M)
FIGURE
6.
Inhibition
by
DEX
and lipocortin
1
of mitogen-induced
PBML
proliferation.
PBML
were stimulated with
PHA-PMA
and
treated with
DEX
(closed triangles)
or
lipocortin
1
(LC
I; open tri-
angles). Proliferation was assessed 3 days after culture initiation;
control cpm values were: background cpm, 404
?
103;
positive
control cpm, 82,679
f
8,629; and ethanol control cpm, 69,844
?
8,379. Data points are the mean of
six
individually performed ex-
periments and indicate the percent suppression, calculated as de-
scribed
in
Fig. 2.
(44), inhibition of IL-I-induced neutrophil migration (43, inhibi-
tion of cellular proliferation (37), and induction of cellular differ-
entiation (36, 46). Here we showed that lipocortin I also inhibits
mitogen-induced and OKT3-stimulated (7, 19) cellular prolifera-
tion. Our results clearly demonstrate that GC-mediated inhibition
of eicosanoid production was the result
of
induction of lipocortins,
as has also been reported in bronchial alveolar lavage cells (47), in
A549 lung adenocarcinoma cells (37, 48), and in differentiated
U-937 cells (34). However, GC-mediated antiproliferative effects
were not exclusively due to enhanced lipocortin expression, since
anti-lipocortin I and anti-lipocortin
II
mAb, while totally abrogating
the GC-mediated inhibition of eicosanoid production, only partially
antagonized GC-mediated antiproliferative effects.
In
essence, the
GC-mediated inhibition of eicosanoid production appears to be
lipocortin dependent (47, 48), while the antiproliferative effects
follow both lipocortin-dependent and lipocortin-independent
pathways (48, 49).
The role of lipocortins in GC-mediated antiproliferative effects
remains to be determined. Lipocortins, which are phosphorylated
on tyrosine, serine, and threonine residues by src-like kinases (50-
52),
may block the action of certain elements in the signaling path-
way that operate as a consequence
of
cellular stimulation, hence
leading to decreased cellular proliferation. In addition to partially
acting via lipocortins, GCs may exert their antiproliferative effects
directly by binding their cytosolic receptor, which, when translo-
cating to the nucleus, binds the promoter region of several cytohne
genes on specific sites collectively referred to as GC response el-
ements (53-55). Binding
of
GC receptor complex to GC response
element DNA sites inhibits cellular proliferation through blockade
of cytokine gene expression at the transcriptional and transcrip-
tional levels in
cis-
or
trans-acting fashions, as previously re-
ported (54, 56).
In conclusion, the demonstration that lipocortins, while mediat-
ing GC effects in inhibiting the release of AA release and eico-
sanoid production, only partially mediate GC-associated antipro-
liferative effects is in line with our earlier thesis (7, 19, 53) that
on March 10, 2010 www.jimmunol.orgDownloaded from
The
Journal
of
Immunology
'''1
A
5237
FIGURE
7.
Inhibition of eicosanoid pro-
duction by NDCA and ETYA. RIA of PCE,
(A)
and LTB,
(6)
levels in the culture super-
natants of PBML stimulated with PHA-PMA
and treated with NDGA (closed circles) or
ETYA (open circles). Data points represent
the mean of five individually performed ex-
periments and indicate the percent suppres-
sion, calculated as described in Figure
5.
4-
C
Q,
Q,
2
n
1001
80-
60-
100
80
60
40
20
Concentration
(M)
log
Concentration
(M)
FIGURE
8.
Failure of NDCA and ETYA to inhibit mitogen-induced
proliferation. PBML were stimulated with PHA-PMA and treated with
DEX
(solid squares), ETYA (open squares), or NDCA (open circles).
Data points represent the mean of four individually performed exper-
iments; control cpm values were: background cpm,
621
t
142;
pos-
itive control cpm,
99,552
2
12,241;
and ethanol control cpm,
93,162
t
16,211.
Percent suppression was calculated as described in
Figure
2.
61
H
80
-
60
40
-
20
-
0
~-~;~~~;.b-;~~~o-~o~..ll"~~~~~"'
'
""""
'
"""'
'
-
10.~
10"
Concentration
(MI
FIGURE
9.
Failure of
AA
to abrogate the GC-mediated antiproliferative
effect. PBML were treated with ethanol (open triangle) or
M
DEX
(closed triangle), reconstituted with AA at the indicated concentrations,
stimulated with PHA-PMA, and cultured for
72
h at
37T.
Data points
represent the mean of six individually performed experiments; control
values were: background cpm,
928
?
180;
positive control cpm,
80,774
2
13,473;
and ethanol control cpm,
84,188
?
12,729.
The per-
cent response was calculated according to: [(test cpm
-
background
cpm)/(control cpm
-
background cpm)l
X
100%.
on March 10, 2010 www.jimmunol.orgDownloaded from
PARTIAL MEDIATION
OF
GLUCOCORTICOID EFFECTS
BY
LIPOCORTINS
5238
100
-
8
(0
c
0
g
60
-
Q)
80
-
a
Q)
40
2
c
C
a
20
-
0
PGE
LTB4
FIGURE
10.
Abrogation of DEX-induced suppression
of
eicosanoid
production
by
anti-lipocortin
I
Ab. PGE, (left) and LTB, (right) levels
were determined by RIA in the culture supernatants of PBML stimu-
lated with PHA-PMA and treated with
DEX
at
10”
M
(DEX), DEX
plus
control lgGl
(IgC
Control),
DEX
plus anti-lipocortin
I
Ab
(1
pLg/ml;
Anti-LC
I),
DEX
plus anti-lipocortin
II
Ab (Anti-LC
II),
or
DEX
plus
anti-lipocortin
I
and anti-lipocortin
II
Abs (Anti-LC
1/11),
Data points
represent the mean
of
five individually performed experiments and
indicate the percent response, calculated according to: [(test conc.
-
background conc.)/(control conc.
-
background conc.)]
X
100%.
GCs exert their antiproliferative effects principally by directly tar-
geting cytokine genes. The role
of
lipocortins in partially mediat-
ing the effects
of
GC remains to be determined.
Acknowledgments
The authors thank Dr. Barbara
P.
Wallner and Dr. Jeoffrey Browning at
Biogen (Cambridge, MA)
for
providing lipocortin
1
and
2
cDNA probes
and anti-lipocortin
I,
11,
and V mAb. The expert technical assistance
of
Edward T. Hadro, Joumana W. Assi, and Dagmara
M.
Chudzik is greatly
appreciated.
References
1.
2.
3.
4.
5.
6.
7.
8
9
10
11
Nagai, H.,
T.
Takizawa, N. Inagaki, T. Sakamoto, T. Shimazawa, and A. Koda.
1989. Anti-allergic action of glucocorticoids.
11.
Effect of glucocorticoids
on
cell-
mediated (type
IV)
allergic reactions.
Arerugi
38:493.
Cupps, T. R., and A. Fauci. 1982. Glucocorticoid-mediated immunoregulation in
man.
lmmunol. Rev.
65:133.
Daeron,
M.,
A.
R.
Sterk, F. Hirata, and T. Ishizaka. 1982. Biochemical analysis
of
glucocorticoid-induced inhibition
of
IgE-mediated histamine release from
mouse mast cells.
J.
lmmunol.
129:1212.
Dennis, G., C.
H.
June,
J.
Mizuguchi,
J.
Ohara, K. Witherspoon, F. D. Finkelman,
V. McMillan, and
J. J.
Mond. 1987. Glucocorticoids suppress calcium mobili-
zation and phospholipid hydrolysis in anti-Ig antibody-stimulated
B
cells.
J.
Im-
munol.
139:2516.
Johnston, D., E. R. Matthews, and G. Melnykovych. 1980. Glucocorticoid effects
creased sphingomyelin synthesis.
Endocrinology
107:1482.
on lipid metabolism in
HeLa cells: inhibition of cholesterol synthesis and in-
Ramachdndran, C. K., and G. Melnykovych. 1983. Transient changes in phos-
pholipid methylation induced by dexamethasone on lymphoid cells.
Cancer Res.
43:5725.
Almawi, W.
Y.,
M.
L.
Lipman, A. C. Stevens, B. Zanker, E. Hadro, and T. B.
proliferation by the synergistic action of
IL-I,
IL-6,
and IFN-y.
J.
Immunol.
Strom. 1991. Abrogation
of
glucocorticosteroid-mediated
inhibition of T cell
146:3523.
Gessaui,
S., S.
McCandless, and
S.
Baglioni. 1988. The glucocorticoid dexa-
J.
Bid.
Chem.
263:7454.
methasone inhibits synthesis of interferon by decreasing the level of its mRNA.
transcriptional and post-transcriptional expression of interleukin 1 in U937 cells.
Knudsen, P.
J.,
C. A. Dinarello, and T. B. Strom. 1987. Glucocorticoids inhibit
J.
Immunol.
139:4129.
Hirata, F.,
M.
L.
Stracke, and E. Schiffmann. 1987. Regulation of prostaglandin
J.
Steroid Biochem.
27:1053.
formation by glucocorticoids and their second messenger, lipocortins.
Wallner, B. P., R. J. Mattaliano, C. Hession, R. L. Cate,
R.
Tizard,
L. K.
Sinclair,
C. Foeller,
E.
P. Chow, J.
L.
Browning,
K.
L.
Ramachandran, and
R.
B. Pepinsky.
with potential anti-inflammatory activity.
Nature
320:77.
1986. Cloning and expression
of
human lipocortin,
a
phospholipase
A2
inhibitor
100
80
8
v)
C
0
,”
60
d)
U
c
40
Y
a
Q,
20
n
PHA/PM4
DEX
IgC
Cont. anti-LC
1
anti-LC
II
anti-LC
1/11
FIGURE
11.
Partial abrogation of DEX-mediated antiproliferative ef-
fects by anti-lipocortin Ab. PBML were stimulated with PHA-PMA and
treated with ethanol (PHNPMA),
1
O-’
M
DEX (DEX), DEX
plus
control
lgGl
(IgG
Cont.),
DEX
plus anti-lipocortin 1 Ab (anti-LC
I),
DEX
plus
anti-lipocortin
II
Ab (anti-LC
II),
or
DEX
plus anti-lipocortin
I
and anti-
lipocortin
II
Abs (anti-LC
1/11),
Data points represent the mean of eight
individually performed experiments; control values were background
cprn, 503
i-
120; positive control cprn, 82,502
?
12,384; and ethanol
control cpm, 80,774
IT
10,008. The percent response was calculated
as described in Figure
9.
12.
Flower, R.
J.,
and G. J. Blackwell. 1979. Anti-inflammatory steroids induce bio-
synthesis of
a
phospholipase A2 inhibitor which prevents prostaglandin genera-
tion.
Nature
278:456.
13.
Peers,
S.
H., and
R.
J.
Flower. 1990. The role of lipocortin in corticosteroid
14. Wahl, L. M., and
L. L.
Lampel. 1987. Regulation
of
human peripheral blood
actions.
Am. Rev. Respir.
Dis.
141:S18.
monocyte collagenase by prostaglandins and anti-inflammatory drugs.
Cell. lm-
15. Webb, D.
S.,
and
I.
A. Roth. 1987. Relationship of glucocorticoid suppression
of
munol.
105:411.
Biol.
41:156.
arachidonic acid metabolism to alteration of neutrophil function.
J.
Leukocyte
16. Russo-Marie, F., and D. Duval. 1982. Dexamethasone-induced inhibition of pros-
taglandin production does not result from
a
direct action
on
phospholipase ac-
tivities but is mediated through
a
steroid-inducible factor.
Biochim. Biophys. Acta
712:
I
77.
17. Grove, R. I., W. D. Willis, and
R.
M.
Pratt. 1985. lnhibition of arachidonic acid
metabolism is not involved in dexamethasone-induced growth inhibition of em-
18. Hullin, F.,
P.
Raynal,
J.
M. Ragab-Thomas,
J.
Fauvel, and H. Chap. 1989. Effect
bryonic palatal development.
Prosfaglundins
Leuk.
Med.
I7:85.
of dexamethasone
on
prostaglandin synthesis and
on
lipocortin
status
in human
endothelial cells: inhibition
of
prostaglandin
I2
synthesis occurring without al-
teration of arachidonic acid liberation and of lipocortin synthesis.
.I.
Biol. Chem.
264:3506.
19. Almawi, W.
Y.,
E.
T.
Hadro, and T. B. Strom. 1991. Glucocorticoid-mediated
immunosuppressive effects do
not
involve altering second messenger function.
Transplantation
52: 133.
20. Laemmli,
U.
K.
1970. Cleavage of structural proteins during the assembly
of
the
head of bacteriophage
T.
Nuture
227:680.
21. Blumberg, D. D. 1987. Creating
a
ribonuclease-free environment.
Methods En-
zymol.
152:572.
22. MacDonald, R.
J.,
G.
H. Swift, A.
E.
Przybyla, and
J.
M. Chirgwin. 1987.
Iso-
lation
of RNA using guanidium salts.
Methods Enzymol.
152:219.
23. Holmdahl,
R.,
and L. Jansson. 1988. Estrogen-induced suppression of collagen
estrogen-mediated suppression of T cell immunity.
Bruin Behav. lmmun.
2:123.
arthritis. 111. Adult thymectomy does not affect the course of arthritis or the
24. Seaman, W.
E.,
T. C. Merigan,
and
N. Talal. 1979. Natural killing in estrogen-
treated mice respond poorly to poly
(1.
C.) despite normal stimulation
of
circu-
lating interferon.
J.
lmmunol.
123:2903.
25.
de Groen, P. C. 1988. Cyclosporine, low-density lipoproteins, and cholesterol.
Muyo
Clin.
Proc.
63:1012.
26. Ablin,
R.
J.,
R. A. Ghatti, and P. GUindn. 1978. Effect of estrogen on tumor-
associated immunity in patients with adenocarcinoma
of
the prostate.
Cancer Res.
30:3702.
27. Siiteri, P.
K.,
F.
Febres,
L.
E.
Clemens,
R.
J.
Chang,
B.
Gondos, and D. P. Stites.
munosuppressant?
Ann.
NY Acud. Sci.
286384.
1977. Progesterone and maintenance of pregnancy: is progesterone nature’s im-
28. Clemens,
L.
E., P.
K.
Siiteri, and D.
P.
Stites. 1979. Mechanism
of
immunosup-
J.
Immunol.
122:1978.
pression of progesterone on maternal lymphocyte activation during pregnancy.
29. Aschkenasy, A. 1976. The effects
of
late thymectomy on the immunosuppressive
action of cortisone and testosterone in
rats
immunized to sheep red blood cells.
Acta Huemutol.
56:212.
on March 10, 2010 www.jimmunol.orgDownloaded from
The
Journal
of
Immunology
5239
30. Wyle, F. A., and
J.
Kent. 1977. Immunosuppression by sex steroid hormones: the
effect upon PHA- and PPD-stimulated lymphocytes.
Clin.
Exp. lmmunol.
27.407.
31. Pavia. C., P.
K.
Siiteri,
I.
D. Perlman, and D.
P.
Stites. 1979. Suppression of
murine allogeneic cell interactions by sex hormones.
J.
Reprod. lmmunol.
1:33.
32. Bienkowski, M.
J.,
M. A. Petro, and
L.
J.
Robinson. 1989. Inhibition of throm-
pocortin
I
as the second messenger.
J.
Biol.
Chem.
264:6536.
boxane
A synthesis in UY37 cells by glucocorticoids: lack of evidence for
li-
33. van Voorhis, B.
J.,
0.
J.
Anderson, and
1.
A. Hill. 1989. The effects of
RU
486
on immune function and steroid-induced immunosuppression in vitro.
J.
Clin.
Endocrinol. Metab.
69:1195.
34. Solito,
E.,
G. Raugei, M. Melli, and
L.
Parente. 1991. Dexamethasone induces the
expression of the mRNA of lipocortin
1
and 2 and the release of lipocortin
1
and
35. Wong, W. T.,
S.
C. Frost, and H.
S.
Nick. 1991. Protein-synthesizing-dependent
5
in differentiated. but not undifferentiated U-937 cells.
FEES Lett.
291:238.
36. Violette,
S.
M.,
I.
King,
J.
L. Browning, R. B. Pepinsky,
B.
P.
Wallner, and
induction
of
annexin
I
by glucocorticoids.
Biochem.
J.
275:313.
A. Sartorelli. 1990. Role
of
lipocortin
I
in the glucocorticoid Induction
of
the
37. Croxtall, J. D., and R.
I.
Flower. 1992. Lipocortin
1
mediates dexamethasone-
terminal differentiation of
a
human squamous carcinoma.
J.
Ceii.
Physioi.
142:70.
induced growth arrest of the A549 adenocarcinoma cell line.
Proc. Natl. Acad.
Sci. USA
89:3571.
38. Flower, R. J. 1988. Lipocortin and the mechanism of action of the glucocorti-
39. Smith,
S.,
T. Tetley, A.
Guz,
and R. Flower. 1990. Detection of lipocortin
I
in
coids.
Br.
J.
Pharmacol.
94:987.
human lung lavage fluid: lipocortin degradation as
a
possible proteolytic mech-
anism in the control of inflammatory mediators and inflammation.
Environ.
Health Perspect.
85:135.
40. Ambrose, M. P., C.
L.
Bahns, and
G.
W. Hunninghake. 1992. Lipocortin I pro-
41. Errasfa, M., and F. Russo-Marie. 1989. A purified lipocortin shares the anti-
duction by human alveolar macrophages.
Am.
J.
Respir.
Cell. Mol.
Biol. 6:17.
inflammatory effect
of
glucocorticosteroids in vivo
in
mice.
Br.
J.
Pharmacol.
97:1051.
42. Kozubik. A.,
J.
Hofmanova,
J.
Hola, and
J.
Netikova.
1993.
The effect
of
nor-
dihydro-guaiaretic acid, an inhibitor of prostaglandin and leukotriene biosynthe-
43. Anderson,
K.
M., and
J.
E. Hanis. 1990.
5,8,11,14-Eicosatetraynoic
acid inhibits
sis, on hematopoiesis of gamma irradiated mice.
Exp. Hematol.
21:138.
PC3 DNA synthesis and cellular proliferation, in part due to its 5"lipoxygenase
activity.
Clin.
Physiol. Biochem.
8:308.
44. Maridonneau-Parini,
I.,
M.
Erasfa,
and F. Russo-Marie. 1989. Inhibition of
02-
phages.
J.
Clin.
Invest.
83:1936.
generation by dexamethasone is mimicked by lipocortin I in alveolar macro-
45. Perretti, M., and R.
J.
Flower. 1993. Modulation of IL-1-induced neutrophil mi-
gration by dexamethasone and lipocortin
l:J.
lmmunol.
150.992.
46. Sartorelli, A.
C.,
K.
lshiguro,
C.
King. M. Morin, and M. Reiss. 1986. Mecha-
nisms involved in the induction of malignant cell differentiation.
Adv.
Enzyme
Regul.
25:507.
47. De Caterina,
R.,
R. Sicari,
D.
Giannessi, P.
L.
Paggiaro, P. Paoletti,
G.
Lazzerini,
W. Bemini,
E.
Solito, and
L.
Parenti. 1993. Macrophage-specific eicosanoid syn-
thesis inhibition and lipocortin-1 induction by glucocorticoids.
J.
Appl. Physiol.
75:2368.
48. Newman,
S.
P., R.
J.
Flower, and J. D. Croxtall. 1994. Dexamethasone suppres-
sion of cyclooxygenase 2 expression
is
not mediated by lipocortin-l in A549
cells.
Biochem. Biophys. Res. Commun.
202:931.
49. Beyaert, R., P. Suffys, F. Van Roy, and W. Fiers. 1990. Inhibition by glucocor-
ticoids
of
tumor necrosis factor-mediated cytotoxicity: evidence against hpocor-
tin involvement.
FEBS
Lett.
262.93.
50.
Johnsson, N., and
K.
Weber. 1990. Alkylation of cysteine 82 of
pl
1
abolishes the
complex formation with the tyrosine-protein kinase substrate p36 (annexin 2,
calpactin
1,
lipocortin 2).
J.
Biol. Chem.
265:14464.
51.
Schlaepfer, D. D., and H. T. Haigler. 1988. In vitro protein kinase C phosphor-
ylation sites of placental lipocortin.
Biochemistry
27:4253.
52. Varticovski,
L.,
S.
B. Chahwala,
M.
Whitman.
L.
Cantley,
D.
Schindler,
E.
P.
Chow,
L.
K.
Sinclair, and R. B. Pepinsky. 1988. Location of sites
in
human
lipocortin I that are phosphorylated by protein tyrosine kinases and protein ki-
nases A and C.
Biochemistrv
27.3682.
53. Almawi, W.
Y.,
K.
L. Sewell.
E.
T. Hadro,
B.
Zanker, and T. B. Suom. 1990.
Mode
of
action of the glucocorticosteroids
as
immunosuppressive agents.
Frog.
Leukocyte Biol.
10A:321.
54. Beato, M. 1989. Gene regulation by steroid hormones.
Cell
56:335.
55.
Hollenberg,
S.
M., V. Giguere,
P.
Segui,
and R. M. Evans. 1987. Colocalization
of DNA-binding and transcriptional activation functions in the human glucocor-
ticoid receptor.
Cell
49:39.
56. Vacca, A., M. P. Felli, A.
R.
Farina,
S.
Martinotti, M. Maroder,
1.
Screpanti,
D.
Meco,
E.
Petrangeli,
L.
Frati, and A. Gulino. 1992. Glucocorticoid receptor-
mediated suppression of the interleukin
2
gene expression through impairment of
the cooperativity between nuclear factor of activated T cells and AP-I enhancer
elements.
J.
Exp. Med.
175:637.
on March 10, 2010 www.jimmunol.orgDownloaded from
... To determine whether DXM exhibited anti-viability effects in MDA-MB-231 cells, we incubated cells with 500 nM DXM and analyzed viability using MTS assays. Consistent with previous reports [44,45], DXM (500 nM) significantly inhibited the viability of MDA-MB-231 cells ( Figure 6D). TTP exerts anti-viability functions by destabilizing the mRNAs of genes involved in cell viability [4,5]. ...
... GCs have been reported to inhibit the growth of cells [43][44][45], and TTP also inhibits the growth of cancer cells by down-regulation of ARE-containing genes involved in cell proliferation [9,10,12]. In our study, TTP expression was induced in MDA-MB-231 cells. ...
Article
Full-text available
The mRNA destabilizing factor tristetraprolin (TTP) functions as a tumor suppressor by down-regulating cancer-associated genes. TTP expression is significantly reduced in various cancers, which contributes to cancer processes. Enforced expression of TTP impairs tumorigenesis and abolishes maintenance of the malignant state, emphasizing the need to identify a TTP inducer in cancer cells. To search for novel candidate agents for inducing TTP in cancer cells, we screened a library containing 1019 natural compounds using MCF-7 breast cancer cells transfected with a reporter vector containing the TTP promoter upstream of the luciferase gene. We identified one molecule, of which the enantiomers are betamethasone 21-phosphate (BTM-21-P) and dexamethasone 21-phosphate (BTM-21-P), as a potent inducer of TTP in cancer cells. We confirmed that BTM-21-P, DXM-21-P, and dexamethasone (DXM) induced the expression of TTP in MDA-MB-231 cells in a glucocorticoid receptor (GR)-dependent manner. To identify potential pathways linking BTM-21-P and DXM-21-P to TTP induction, we performed an RNA sequencing-based transcriptome analysis of MDA-MB-231 cells at 3 h after treatment with these compounds. A heat map analysis of FPKM expression showed a similar expression pattern between cells treated with the two compounds. The KEGG pathway analysis results revealed that the upregulated DEGs were strongly associated with several pathways, including the Hippo signaling pathway, PI3K-Akt signaling pathway, FOXO signaling pathway, NF-κB signaling pathway, and p53 signaling pathway. Inhibition of the FOXO pathway using a FOXO1 inhibitor blocked the effects of BTM-21-P and DXM-21-P on the induction of TTP in MDA-MB-231 cells. We found that DXM enhanced the binding of FOXO1 to the TTP promoter in a GR-dependent manner. In conclusion, we identified a natural compound of which the enantiomers are DXM-21-P and BTM-21-P as a potent inducer of TTP in breast cancer cells. We also present new insights into the role of FOXO1 in the DXM-21-P- and BTM-21-P-induced expression of TTP in cancer cells.
... ANX A1 regulates ERK signaling pathway activation and inhibits cyclin D1 expression, therefore reducing cell proliferation (20). Evidence reveals that ANX A1 functions as an inhibitor of signal transduction pathways that leads to cell proliferation in JACRO cells (21) and lymphocytes (31). However, the effects of over-expression of ANX A1 in rapidly proliferating hepatocytes (7), as well as human foreskin fibroblasts (32) indicates that the underlying mechanism is more complex. ...
Article
Full-text available
Annexin A1 (ANX A1) is essential in cell differentiation and proliferation. However, the role of ANX A1 in bone marrow‑derived mesenchymal stem cell (BM‑MSC) osteogenic differentiation and proliferation remains unclear. To investigate whether endogenous ANX A1 influences BM‑MSC proliferation and osteogenic differentiation, a stable ANX A1‑knockdown cell line was generated using short hairpin RNA (shRNA). The proliferation rate of BM‑MSCs was analyzed by 3‑(4,5‑Dimethylthiazol‑2‑yl)‑2,5‑diphenyltetrazolium bromide proliferation assay. Additionally, BM‑MSCs were differentiated into osteoblasts and subsequently used to isolate total proteins to analyze the expression of ANX A1. Cell differentiation was assayed using Alizarin red S staining. The results revealed that the knockdown of ANX A1 in BM‑MSCs exerts no apparent effect on the proliferation rate under normal conditions, however, following exposure to an osteogenic medium, downregulation of ANX A1 protected cells from the effect of osteogenic medium‑induced inhibition of cell proliferation. Silencing ANX A1 with shRNA significantly inhibited the phosphorylation of extracellular signal‑regulated kinase 1/2 and the expression of differentiation‑associated genes (including runt‑related transcription factor 2, osteopontin and osteocalcin) during osteogenesis and resulted in reduced differentiation of BM‑MSCs. The results indicate the potential role of ANX A1 in the regulation of BM-MSC proliferation and osteogenic differentiation.
... The concentration , the dose of the drug and the application method can all be altered according to the response. Corticosteroids have an important role in skin diseases because of their anti-inflammatory [1], immunosuppressive [2], and anti-proliferative effects [3] on the keratinocytes. ...
Article
Full-text available
Background: Corticosteroids are extremely useful in the treatment of inflammatory skin disorders. Topical steroid applications are the most effective treatment for all types of psoriasis. Aims: To compare the efficacy of once daily versus alternate day application of topical steroid clobetasol propionate 0.05% (Tenovate cream®) in patients who have mild to moderate plaque psoriasis. Settings and Design: This study was conducted on 89 patients of plaque psoriasis, who attended the skin OPD in our hospital. Methods and Material: The patients who had a mild to moderate severity of plaque psoriasis were selected. Those patients who required systemic corticosteroids, those who were already undergoing any psoriasis treatment, those who had any other debilitating illness and pregnant and lactating women were excluded from the study. Eighty nine patients were enrolled for the study after taking written informed consent from them and they were randomly allocated into two groups. Two patients dropped out, one in each group and 1 of group 2 was prescribed systemic corticosteroids. Group 1- once daily application (n= 44) Group 2- alternate day application (n= 42) An objective scoring was done on the basis of the PSI (Psoriasis Severity Index) score, which was graded from 0-4. Follow ups were done in the 2nd, 4th and 6th weeks. Statistical Analysis: It was done by the Student’s ‘t’ test and ANOVA. Results and Conclusions: The results showed that there was equal improvement in both the groups in the 2nd week, since the p-value was not significant (P> 0.05), but in the 4th and 6th weeks, there was less improvement in the alternate day group (P< 0.05). An intra group comparison indicated that clobetasol was effective both in the once daily and the alternate day groups, but clinical and symptomatic improvement occurred more quickly in the once daily group after 6 weeks of assessment. Thus, it can be concluded that the alternate day application of the topical steroid clobetasol propionate cream is as effective as the once daily application in the initial 2 weeks, but by 6 weeks, its efficacy decreases. Therefore, initially, we can advocate a less frequent application of potent topical steroids but for the complete remission of the disease, the application frequency should be once daily.
... The concentration , the dose of the drug and the application method can all be altered according to the response. Corticosteroids have an important role in skin diseases because of their anti-inflammatory [1], immunosuppressive [2], and anti-proliferative effects [3] on the keratinocytes. ...
Article
Full-text available
Background: Corticosteroids are extremely useful in the treat-ment of inflammatory skin disorders. Topical steroid applications are the most effective treatment for all types of psoriasis.
Chapter
Psoriasis is a chronic inflammatory skin disease that affects 2–4% of the world’s population. Approximately 80% of psoriasis patients have limited, localized mild-to-moderate disease where topical therapies serve as the mainstay of treatment. Topical therapies can provide both high efficacy as well as safety in this population. Furthermore, in patients with moderate-to-severe disease, short-term topical treatments may provide symptomatic relief, minimize doses of photo- or systemic therapy, and be of benefit for resistant lesions as part of a combination regimen. This chapter provides an evidence-based concise overview of the topical treatments for psoriasis including corticosteroids, vitamin D derivatives, tazarotene, calcineurin inhibitors, anthralin, salicylic acid, coal tar, and lactic acid.
Article
Full-text available
Glucocorticoids (GCs) are used as immunosuppressive and antiinflammatory agents in organ transplantation and in treating autoimmune diseases and inflammatory disorders. GCs were shown to exert their antiproliferative effects directly through blockade of certain elements of an early membrane-associated signal transduction pathway, modulation of the expression of select adhesion molecules, and by suppression of cytokine synthesis and action. GCs may act indirectly by inducing lipocortin synthesis, which in turn, inhibits arachidonic acid release from membrane-bound stores, and also by inducing transforming growth factor (TGF)-β expression that subsequently blocks cytokine synthesis and T cell activation. Furthermore, by preferentially inhibiting the production of Th1 cytokines, GCs may enhance Th2 cell activity and, hence, precipitate a long-lasting state of tolerance through a preferential promotion of a Th2 cytokine-secreting profile. In exerting their antiproliferative effects, GCs influence both transcriptional and posttranscriptional events by binding their cytosolic receptor (GR), which subsequently binds the promoter region of cytokine genes on select DNA sites compatible with the GCs responsible elements (GRE) motif. In addition to direct DNA binding, GCs may also directly bind to, and hence antagonize, nuclear factors required for efficient gene expression, thereby markedly reducing transcriptional rate. The pleiotrophy of the GCs action, coupled with the diverse experimental conditions employed in assessing the GCs effects, indicate that GCs may utilize more than one mechanism in inhibiting T cell activation, and warrant careful scrutiny in assigning a mechanism by which GCs exert their antiproliferative effects. © 1998 Elsevier Science Inc.
Article
Glucocorticoids (GCs) exert their potent anti-inflammatory and immunosuppressive effects through an intricate combination of mechanisms. Through both pre- and post-transcriptional means, GCs modify gene regulation in target cells by interacting with the cytosolic glucocorticoid receptor (GR). Among their actions, GCs inhibit the production of pro-inflammatory cytokines. This inhibition is accomplished by antagonism of proinflammatory transcription factors by either the GR itself or by de novo-synthesised antagonists such as glucocorticoid-induced leucine zipper and inhibitor of κB (IκB). Preferential inhibition of type-1 cytokines leads to an eventual shift towards a Th2 profile among CD4+ T-lymphocytes, reducing the pro-inflammatory Th1 population. Not only are pro-inflammatory cytokines inhibited, but the effects of these cytokines upon target cells are diminished due to GC-mediated interference with cytokine receptor signalling. This leads to increases in T-cell, thymocyte, and eosinophil apoptosis, reduced T-cell activation, and decreased production of nitric oxide. Further to these effects, GCs inhibit prostaglandin and leukotriene production by inducing synthesis of lipocortin-1. GCs also up-regulate expression of the anti-inflammatory cytokine transforming growth factor-β (TGF-β) in certain cells. A further mechanism by which GCs suppress normal inflammatory responses is by down-modulating adhesion molecules on antigen-presenting cells (APCs). It is this synergy of many effects that accounts for the potency of GC action and therefore for the utility of these drugs, although it is this very complexity that hampers study in this area. Consideration of GC mechanisms of action is important in order to develop an understanding of the long-term effects of use of these heavily prescribed but poorly understood drugs.
Article
Glucocorticoid drugs are frequently used nonspecifically by veterinarians to control clinical signs associated with central nervous system disease. However, this use is infrequently justified and can also be associated with detrimental long-term patient outcomes. First, there are few diseases for which glucocorticoids are the preferred or definitive treatment. Second, their actions may blunt subsequent diagnostic efforts, for instance, by altering MRI appearance or cerebrospinal fluid cell content, or lead owners to abandon pursuit of more appropriate therapies if they perceive the first-line steroid therapy to be a failure.
Article
Purpose: We have identified annexin II mRNA expression in the rat cornea and demonstrated immunolocalization of annexin II in normal and injured corneas. Furthermore, to investigate possible interaction between annexin II and its extracellular ligand tenascin during corneal wound healing, we also examined tenascin expression simultaneously. Methods: Total RNA was extracted from the corneal tissue of male Wistar rats as well as from the cell cultures of corneal epithelial cells and keratocytes. cDNA was obtained by reverse transcription (RT). Annexin II mRNA expression was examined by polymerase chain reaction (PCR). Western blot analysis of annexin II protein was performed with protein samples that were obtained from the corneal tissue and cell cultures. In addition, the localization of annexin II and that of tenascin were clarified by immunohistochemical analysis, using both uninjured and epithelial scraped corneas. Results: RT-PCR analysis revealed that annexin II mRNA was expressed in corneal tissues, epithelial cells and keratocytes. Western blot analysis of corneal epithelium and keratocytes showed a 38kDa band that corresponded to the molecular weight of annexin II. Immunohistochemical study showed that annexin II was present in keratocytes as well as the basal cells of corneal epithelium in the central cornea, and basal/suprabasal cells of the limbal epithelium. Positive annexin II immunoreactivity is translocated from cytoplasm to the cell periphery/extracellular position during the epithelial wound healing process. Annexin II and tenascin are coexpressed on the basal surface of limbal epithelium and the leading edge of the healing epithelium. The reappearance of cytoplasmic annexin II staining at the periphery of the cornea correlated with the epithelial cell proliferation. Conclusions: Annexin II is abundantly expressed in corneal tissue. The translocalization of annexin II protein suggests its role in the corneal epithelial migration during wound healing. The colocalization of annexin II and tenascin in migrating and proliferating corneal epithelial cells suggests that annexin II-tenascin interaction may play a role in epithelial wound healing.
Article
Full-text available
Tumor-associated antigen-induced leukocyte adherence inhibition has been used as an in vitro criterion for evaluating the effect of estrogen on cell-mediated antitumor-associated immunity in patients with adenocarcinoma of the prostate. Significant (p less than 0.05) suppression from the reactivity obtained with untreated patients' leukocytes to allogeneic extracts of malignant prostatic tissue ranging from 19 to 80% was observed in all patients following preincubation of their leukocytes with diethylstilbesterol diphosphate. The observed suppression of tumor-associated immunity in the presence of exogenous estrogen provides further evidence to earlier studies that demonstrated estrogenic suppression of nonspecific cellular responsiveness as evaluated by phytohemagglutinin-induced lymphocytic blastogenesis of normal and prostatic cancer patients' lymphocytes and for the initially suggested concern over the efficacy of estrogenic therapy and its adverse effect on host cell-mediated immunological responsiveness.
Article
Full-text available
The immunosuppressant hormone dexamethasone (Dex) interferes with T cell-specific signals activating the enhancer sequences directing interleukin 2 (IL-2) transcription. We report that the Dex-dependent downregulation of 12-O-tetradecanoyl-phorbol-13-acetate (TPA) and calcium ionophore-induced activity of the IL-2 enhancer are mediated by glucocorticoid receptor (GR) via a process that requires intact NH2- and COOH-terminal and DNA-binding domains. Functional analysis of chloramphenicol acetyltransferase (CAT) vectors containing internal deletions of the -317 to +47 bp IL-2 enhancer showed that the GR-responsive elements mapped to regions containing nuclear factor of activated T cells protein (NFAT) (-279 to -263 bp) and AP-1 (-160 to -150 bp) motifs. The AP-1 motif binds TPA and calcium ionophore-induced nuclear factor(s) containing fos protein. TPA and calcium ionophore-induced transcriptional activation of homo-oligomers of the NFAT element were not inhibited by Dex, while AP-1 motif concatemers were not stimulated by TPA and calcium ionophore. When combined, NFAT and AP-1 motifs significantly synergized in directing CAT transcription. Such a synergism was impaired by specific mutations affecting the trans-acting factor binding to either NFAT or AP-1 motifs. In spite of the lack of hormone regulation of isolated cis elements, TPA/calcium ionophore-mediated activation of CAT vectors containing a combination of the NFAT and the AP-1 motifs became suppressible by Dex. Our results show that the IL-2-AP-1 motif confers GR sensitivity to a flanking region containing a NFAT element and suggest that synergistic cooperativity between the NFAT and AP-1 sites allows GR to mediate the Dex inhibition of IL-2 gene transcription. Therefore, a Dex-modulated second level of IL-2 enhancer regulation, based on a combinatorial modular interplay, appears to be present.
Article
Cancer appears to be a disease of altered maturation, with changes in genetic expression leading to a situation in which the physiological regulation of cellular proliferation and maturation are altered. Environmental factors as well as defined chemical agents have been demonstrated to have the capacity to convert neoplastic cells to end-stage forms with a finite life span through a process characteristic of cellular maturation. The correction of genetic defects by these inducers of differentiation does not appear to be required; the critical feature is that the differentiated cells assume a state in which they no longer possess the capability for continued cellular replication. The extrapolation of these advances, accomplished in experimental systems, to clinical practice should yield significant decreases in the neoplastic cell burden without the degree of morbidity produced by aggressive therapy with cytodestructive agents, especially when employed in multidrug combinations. The ultimate introduction of differentiation as a therapeutic approach to cancer treatment if attained, however, will require a variety of principles to be established, so that (a) optimum efficacy may be obtained from each agent, (b) the fabrication of new agents with major changes in the ratio of the concentrations required to produce cytotoxicity relative to those necessary to initiate maturation is attained, and (c) the elucidation of non-antagonistic combinations of differentiation inducing agents with or without cytotoxic drugs is achieved to combat the problem of tumor cell heterogeneity.
Article
Investigations have been carried out on the action of several steroid hormones on lymphocyte functions in inbred strains of mice. The recognitive, proliferative and effector phases of allogeneic cell interactions in vitro were assessed using a mixed lymphocyte reaction (MLR) and cell-mediated lympholysis (CML). In MLR containing Balb/c (responder) and C57bl/6 (stimulator) splenocytes DNA synthesis was markedly reduced in the presence of progesterone, cortisol or estradiol. In CML, progesterone and estradiol (1-5 microgram/ml) blocked in vitro generation of cytotoxic lymphocytes, while cultures with cortisol were partially inhibited. None of these hormones suppressed the cytotoxic activity of previously sensitized effector cells generated in vitro. Cultures containing testosterone expressed both normal DNA synthesis in MLR and cytotoxic activity in the CML test. These findings suggest a selective pattern of immunosuppression by sex hormones which may be important in preventing graft rejection or graft-versus-host interactions which may arise as a consequence of fetal engraftment during pregnancy.
Article
Natural killing by mouse spleen cells can be stimulated in vivo by interferon or by agents that stimulate interferon, such as poly I.C. Natural killing can be suppressed in vivo by the sustained administration of 17 beta-estradiol. In BALB/c mice that had been treated with 17 beta-estradiol for 10 weeks, natural killing did not respond to intravenous poly I.C, although stimulation of circulating interferon was equal to controls. Estradiol, then, does not block interferon production but does suppress the response of natural killer cells to interferon. It is suggested that estrogens either block the maturation of natural killer cells or reduce the number of natural killer cell precursors.
Article
ASPIRIN prevents prostaglandin (PG) generation by directly inhibiting the cyclo-oxygenase enzyme responsible for PG biosynthesis1-3. In addition, there is now conclusive evidence that anti-inflammatory steroids can also prevent PG generation4-13. Unlike the aspirin-like drugs, steroids have no anti-cyclo-oxygenase activity but exert their action by preventing the release from phospholipids of the fatty acid substrates required for PG biosynthesis4-9,12,13. We have shown that stimulation of thromboxane A2 (TXA2) release by agents such as histamine, 5-hydroxytryptamine and rabbit aorta contracting substance-releasing factor (RCS-RF) (but not arachidonic acid) is inhibited by anti-inflammatory steroids, and that their potency in this action closely parallels their anti-inflammatory activity12,13. Furthermore, their mechanism of action involves the inhibition of phospholipase A2 activity, and thus of arachidonate release within the lung12,13. In several other tissues, the mechanism of steroid hormone action depends on the combination of thesteroid with a cytosolic receptor protein, the translocation of this drug-receptor complex to the nucleus and the initiation of protein biosynthesis14-16. We now show that similar events occur in the guinea pig perfused lung before inhibition by steroids of phospholipase A2 activity (and thus TXA2 generation). We have discovered a steroid-induced factor which mimics the anti-phospholipase effects of these anti-inflammatory agents.
Article
The immunological effects of cortisone and testosterone were studied in intact and lately thymectomized rats immunized to sheep red cells. The results showed that the effects of low doses of cortisone as well as those of high doses of testosterone (drop in the total number of rosette- and plaque-forming cells: RFC and PFC) were conditioned by the involution of the thymic cortex provoked by such hormonal doses, since these changes did not occur in thymectomized rats. The effects of high doses of cortisone were partly not influenced by thymectomy (drop in the RFC and PFC levels) but partly thymus-dependent (drop in the serum antibody titers). The latter were apparently related to advanced lesions of the thymic medulla.
Article
Progesterone, estradiol, testosterone, cortisol, and 11-desoxycortisol (compound S) were added to cultures of human lymphocytes stimulated with phytohaemagglutinin (PHA) and purified protein derivative (PPD). The immunosuppressive effect of cortisol was verified and the three sex-steroid hormones also were found to inhibit lymphocyte transformation although at concentrations higher than for cortisol. Compound S, a steroid of low biological potency, also had immunosuppressive activity. At concentrations (0-01-1-0 microng/ml), progesterone, oestrogen, testosterone, and Compound S augmented the transformation response to PPD but not to PHA. Marked variation from individual to individual in the suppressive effects of all the steroids were noted. The clinical implications of immunosuppression by the sex steroid hormones are discussed.
Article
The synthetic glucocorticoid dexamethasone (1 microM to 1 pM) strongly (maximum greater than 80%) inhibits proliferation of the A549 human lung adenocarcinoma line (EC50 greater than 1 nM) and leads to the appearance, or a further increase (approximately 3-fold) in the expression on the cell surface, of the calcium and phospholipid binding protein lipocortin (annexin) 1. Both these effects, which are shared by hydrocortisone (1 microM) but not by progesterone or aldosterone (1 microM), are inhibited by the antiglucocorticoids RU38486 and RU43044 (1 microM). The nonsteroidal antiinflammatory drugs indomethacin (1 microM) and naproxen (10 microM) and human recombinant lipocortin 1 (0.05-5.0 micrograms/ml) also produce growth arrest in this cell line. During proliferation A549 cells spontaneously release prostaglandin E2 [10-20 ng (28-57 pmol) per ml per 5-day period] into the growth medium. In concentrations that cause growth-arrest, dexamethasone, indomethacin, and lipocortin 1 abolish the generation of this eicosanoid by A549 cells. Prostaglandin E2 itself (0.01-1 pM) stimulates cell growth and partially reverses (approximately 50%) the inhibition of growth caused by dexamethasone and indomethacin. Addition of the neutralizing anti-lipocortin 1 monoclonal antibody 1A (5 micrograms/ml), but not the nonneutralizing anti-lipocortin monoclonal antibody 1B, substantially reversed (greater than 80%) the inhibitory activity of dexamethasone on both growth and prostaglandin E2 synthesis. The generation of prostaglandin E2 by A549 cells seems to be an important regulator of cell proliferation in vitro and the dexamethasone-induced suppression of proliferation in this model is attributable to eicosanoid inhibition caused by lipocortin 1.