ArticlePDF Available

Tumor Necrosis Factor Induces Lipopolysaccharide Tolerance in a Human Adenocarcinoma Cell Line Mainly through the TNF p55 Receptor

Authors:

Abstract and Figures

This study demonstrates that lipopolysaccharide (LPS) mediates induction of transcription factor NF kappa B and activation of the cytomegalovirus (CMV) promoter-enhancer in the SW480 cell line. These cells do not express a functional membrane CD14. The LPS response in SW480 cells was weaker and markedly slower than the tumor necrosis factor (TNF) response. Pretreatment with TNF for 72 h inhibited both TNF, tumor necrosis factor receptor (TNFR) p55, TNFR p75, and LPS-mediated activation of nuclear factor -kappa B (NF kappa B), whereas pretreatment with LPS only inhibited the LPS response. TNFR p55 antibody pretreatment resulted in marked inhibition of the LPS response, while pretreatment with TNFR p75 antiserum only had a weak inhibitory effect. Flowcytometric analysis showed that LPS binding as well as expression of TNFR p55 and TNFR p75 were not affected by LPS or TNF pretreatment, indicating that the observed inhibition is not due to reduction of specific binding sites at the cell surface. The results suggest that LPS signaling in SW480 cells involves intracellular components which may be depleted or inactivated via TNFR p55, indicating that the LPS and TNFR p55 pathways overlap. We propose that TNFR p55 can mediate activation of NF kappa B and cytomegalovirus promoter-enhancer in SW480 cells via two distinct mechanisms, one which is activated only via TNFR p55 and leads to rapid activation of NF kappa B, and another which is overlapping with the LPS pathway.
Content may be subject to copyright.
Tumor Necrosis Factor Induces Lipopolysaccharide Tolerance
in a Human Adenocarcinoma Cell Line Mainly through the
TNF p55 Receptor*
(Received for publication, March 29, 1995, and in revised form, July 9, 1995)
Astrid Lægreid‡§, Liv Thommesen§, Tove Gullstein Jahr‡, Anders Sundan‡, and Terje Espevik‡
From the Institute of Cancer Research and Molecular Biology and §UNIGEN, Center for Molecular Biology,
University of Trondheim, N-7005 Trondheim, Norway
This study demonstrates that lipopolysaccharide
(LPS) mediates induction of transcription factor NF
k
B
and activation of the cytomegalovirus (CMV) promoter-
enhancer in the SW480 cell line. These cells do not ex-
press a functional membrane CD14. The LPS response in
SW480 cells was weaker and markedly slower than the
tumor necrosis factor (TNF) response. Pretreatment
with TNF for 72 h inhibited both TNF, tumor necrosis
factor receptor (TNFR) p55, TNFR p75, and LPS-medi-
ated activation of nuclear factor -
k
B (NF
k
B), whereas
pretreatment with LPS only inhibited the LPS response.
TNFR p55 antibody pretreatment resulted in marked
inhibition of the LPS response, while pretreatment with
TNFR p75 antiserum only had a weak inhibitory effect.
Flowcytometric analysis showed that LPS binding as
well as expression of TNFR p55 and TNFR p75 were not
affected by LPS or TNF pretreatment, indicating that
the observed inhibition is not due to reduction of spe-
cific binding sites at the cell surface. The results suggest
that LPS signaling in SW480 cells involves intracellular
components which may be depleted or inactivated via
TNFR p55, indicating that the LPS and TNFR p55 path-
ways overlap. We propose that TNFR p55 can mediate
activation of NF
k
B and cytomegalovirus promoter-en-
hancer in SW480 cells via two distinct mechanisms, one
which is activated only via TNFR p55 and leads to rapid
activation of NF
k
B, and another which is overlapping
with the LPS pathway.
Lipopolysaccharide (LPS),
1
a major membrane component of
Gram-negative bacteria, plays an important role in the patho-
genesis of Gram-negative sepsis leading to septic shock (1).
LPS is a potent stimulator of monocytes and macrophages
which respond by production of tumor necrosis factor (TNF),
interleukin (IL)-1, IL-6, IL-8, eicosanoids (2), and nitric oxide
(3). LPS activates monocytes and macrophages via CD14, a
glycosyl-phosphatidylinositol-anchored surface protein (4).
However, LPS receptors other than CD14 may also contribute
to LPS signaling (5–7).
A wide variety of other cell types are also affected by LPS,
and some of these cells do not express membrane CD14. Thus,
LPS has been reported to stimulate arachidonate metabolism
and surface expression of adhesion molecules in endothelial
cells, and it can induce aggregation of platelets, stimulate
cytokine release from mast cells and fibroblasts, and lead to
generation of chemotactic factors in epithelial cells (8). Al-
though CD14 is not present on the plasma membrane of these
cells, soluble (s)CD14 present in serum is essential for their
stimulation by LPS (9, 10).
Transcription factors activated by LPS include NF
k
B (10, 11)
and NF-IL6 (12, 13). TNF is also known as a potent inducer of
NF
k
B as well as of other transcription factors including AP-1,
NF-IL6, IRF-1, and NF-GMa (14). NF
k
B belongs to the rel
family of transcription factors which form a number of different
hetero- and homodimers participating in the regulation of a
large number of genes involved in the immune response (15).
NF
k
B proteins are constitutively expressed in the cytoplasm,
bound to inhibitor I
k
B, and are released and translocated to the
nucleus upon phosphorylation and degradation of I
k
B (16–18).
Most likely, more than one I
k
B kinase is involved, and different
stimuli lead to phosphorylation and degradation of different
I
k
B species. Thus, while I
k
B-
a
is degraded both by TNF and
LPS, I
k
B-
b
is only degraded in response to LPS and IL-1, but
not in response to TNF (19).
TNF can induce NF
k
B via two different receptors, TNFR p55
and TNFR p75 (20). In most cells, TNFR p55 is responsible for
activation of NF
k
B (21, 22), and recent evidence indicates that
TNFR p55 signaling involves ceramide (23, 24). In some cell
types, NF
k
B may also be activated by TNFR p75 (20), but
TNFR p75 signaling mechanisms are still poorly understood.
Association of serine/threonine kinases with cytosolic domains
of both TNFR p75 (25) and TNFR p55 (26, 27) have been
reported, and recently, two cytosolic proteins which specifically
associate with TNFR p75 intracellular domains were identified
and cloned (28). However, no intracellular pathways activated
by any of these proteins have yet been identified, and it is not
known whether they are involved in TNF-mediated activation
of NF
k
B.
LPS-mediated activation of NF
k
B in monocytes is dependent
on intracellular protein tyrosine kinase activity (29). Similarly,
TNF-mediated activation of NF
k
B in lysates of the monocyte-
derived cell line U937 was inhibited by a tyrosine kinase in-
hibitor (30). Also, both LPS- and TNF-induced activation of
NF
k
B is inhibited by the antioxidant pyrrolidine dithiocarba-
mate (31, 32) indicating that reactive oxygen intermediates
play a role in both LPS- and TNF-mediated NF
k
B activation.
So far, LPS signal transduction has been most extensively
studied in cells expressing a functional membrane CD14. How-
ever, LPS effects on CD14 negative cell types may also contrib-
* This work was supported by The Norwegian Cancer Society and by
The Research Council of Norway and Pronova Biopolymer. 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 1734 solely to indicate this fact.
To whom correspondence should be addressed: Institute of Cancer
Research and Molecular Biology, University of Trondheim, N-7005
Trondheim, Norway. Tel.: 47-73-59-86-68; Fax: 47-73-59-88-01.
1
The abbreviations used are: LPS, lipopolysaccharide;
b
-gal,
b
-galac-
tosidase; CMV, cytomegalovirus; mAb, monoclonal antibody; PC-PLC,
phosphatidyl-specific phospholipase C; PMA, phorbol 12-myristate 13-
acetate; p75 AS, TNFR p75 antiserum; TNF, tumor necrosis factor;
TNFR tumor necrosis factor receptor; IL, interleukin; FITC, fluorescein
isothiocyanate; LBP, LPS-binding protein.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 270, No. 43, Issue of October 27, pp. 25418–25425, 1995
© 1995 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
25418
by guest on November 13, 2016http://www.jbc.org/Downloaded from
ute to LPS-mediated pathology. In the present study, we show
that LPS activates the transcription factor NF
k
B and the cy-
tomegalovirus (CMV) promoter-enhancer in SW480 human ad-
enocarcinoma cells which do not have a functional membrane
CD14. Since both TNFR p55 and TNFR p75 independently
activate SW480 cells (20), we considered it important to com-
pare the LPS signaling mechanism to the signal transduction
pathways activated by the TNF receptors. The results demon-
strate that TNF induces LPS tolerance in SW480 cells and that
TNFR p55 and LPS may activate overlapping pathways.
MATERIALS AND METHODS
Cell Cultivation and Stimulation—Human colon adenocarcinoma
cells, SW480/
b
-gal (generously provided by Dr. Gerald Ranges, Miles
Inc., West Haven, CT), contain a
b
-galactosidase (
b
-gal) gene under the
control of the CMV immediate early promoter-enhancer (33). SW480/
b
-gal were grown in RPMI 1640 (Life Technologies, Inc. Laboratories,
Paisley, Scotland), supplemented with 2 mML-glutamine, 10% heat-
inactivated fetal calf serum (HyClone, Logan, UT), and 40
m
g/ml gara-
mycin (fetal calf serum medium). Stimulation with LPS was routinely
carried out in RPMI 1640 medium supplemented with glutamine, 20%
human A
1
serum (The Blood Bank, University Hospital of Trondheim,
Trondheim, Norway), and garamycin (A
1
medium). Experiments con-
ducted at serum-free conditions were performed in AIM medium (Life
Technologies, Inc.).
Reagents—LPS from smooth Salmonella minnesota 6261 and from
rough S. minnesota Re595 (Sigma) as well as S. minnesota lipid A di-
and monophosphate (Sigma) were solubilized in 0.9% NaCl at stock
solutions of 2 mg/ml (LPS) or 1 mg/ml (lipid A). Recombinant (r)CD14
and (r)LBP were generously provided by Dr. M. Lichenstein and Dr. M.
Zukowski, AMGEN (Thousand Oaks, CA) and were prepared as de-
scribed (34). Human recombinant TNF, with specific activity 7.6 310
7
units/mg protein, was generously supplied by Dr. Refaat Shalaby,
Genentech Inc. (South San Francisco, CA). Anti-CD14 monoclonal an-
tibody (mAb) 3C10 was obtained from a subclone of the hybridoma
ATCC TIB 228 (American Type Culture Collection).
TNFR p75 antiserum (p75 AS) was generated by multiple injections
of a rabbit with recombinant soluble TNFR p75 (20). The mAb htr-9
against TNFR p55 (35) was generously provided by Dr. M. Brockhaus,
Hoffmann La-Roche Ltd. (Basel, Switzerland). Biotinylation of htr-9
and mAb utr-4 against TNFR p75 (35) was performed as described (36).
The mAb 6H8 directed against a widely distributed 180-kDa membrane
protein
2
was used as a control antibody. All mAbs were purified on a
Sepharose goat anti-mouse IgG column (Zymed Laboratories Inc.,
South San Francisco, CA). Benzamidine (Sigma) was dissolved in 50%
ethanol at 0.5 Mand phenylmethylsulfonyl fluoride (Sigma) in isopropyl
alcohol at 0.1 M.
b
-Galactosidase Assay—The
b
-galactosidase assay was performed
essentially as described previously (20). Substrate conversion was
measured as optical density (OD) at 570 nm. For pretreatment studies,
cells were seeded out in A
1
medium containing pretreatment reagents.
After 72 h, the plates were washed three times in Hanks’ buffered salt
solution, and test reagents were added in A
1
medium.
Quantitative Band Shift Assays—Preparation of nuclear extracts
and band shift analysis was performed essentially as described (20).
Briefly, equal amounts of nuclear protein from each sample were incu-
bated with 1
m
g of poly(dI-dC) (Pharmacia Fine Chemicals, Uppsala,
Sweden), in binding buffer (20 mMHEPES, pH 7.9, 50 mMKCl, 1 mM
EDTA, 1 mMdithiothreitol, 0.25 mg/ml bovine serum albumin, 2%
Ficoll) (20
m
l final volume) for 10 min at room temperature. Then, 17
fmol (1–5 310
4
counts/min) of end-labeled NF
k
B-specific double-
stranded oligonucleotide probe 59-AGTTGAGGGGACTTTCCCAGG-39
(Promega Corp., Madison, WI) was added, and the mixture was incu-
bated for another 15–20 min. The samples were applied on native
polyacrylamide gels (7% acrylamide, 0.25 3Tris borate-EDTA, 2.5%
glycerol) and run at 100 V for 1 h and then at 130 V for 2–3 h, after
which the gels were dried and exposed to x-ray film (X-Omat AR,
Kodak, Rochester, NY) for 2–16 h. Specific band shifts were quantitated
with a PhosphoImager (Molecular Dynamics, Sunnyvale, CA) by meas-
uring the radioactivity within a rectangle enclosing the band shift in gel
scans after exposures of 18–24 h. Radioactivity counts, which are a
2
B. Naume, A. Sundan, and T. Espevik, unpublished results.
FIG.1. Induction of
b
-galactosidase activity in SW480/
b
-gal
cells by LPS from S.minnesota,E.coli or Pseudomonas,orbyS.
minnesota lipid A.
b
-Galactosidase activity was measured after stim-
ulation of the cells for4hinA
1
medium. Results (mean values of
duplicates) of a representative experiment are given.
FIG.2.Serum dependence and effect of CD14 mAb, rCD14, and rLBP on Re595 LPS-induced
b
-galactosidase activity. SW480/
b
-gal
cells were stimulated for 4 h with Re595 LPS in medium with increasing amounts of human serum (A), in A
1
medium (20% human serum) with
or without neutralizing CD14 mAb 3C10 or control mAb 6H8 (B), or in serum-free AIM medium with or without the addition of recombinant CD14,
recombinant LBP or 20% human serum (C). Results (mean values of duplicates) of a representative experiment are given.
Comparison between LPS and TNF Signal Transduction 25419
by guest on November 13, 2016http://www.jbc.org/Downloaded from
function of both gel exposure times and of the specific activity of the
probe, were termed “PhosphoImager units” and were used to compare
the relative amounts of radioactivity in band shifts within one gel. In
order to control that a similar amount of nuclear proteins was applied
for each sample, we (i) performed band shift assays with
32
P-labeled
OCT oligonucleotide (Promega) (59-TGTCGAATGCAAATCACTAGAA-
39) which binds to constitutively expressed “octamer” proteins (37), or
(ii) measured the intensity of nonspecific bands in the NF
k
B band shift.
All samples were analyzed in at least two band shift assays.
Flow Cytometric Analysis of TNFR mAb and FITC-LPS Binding—
SW480/
b
-gal cells were pretreated with 10 ng/ml TNF or with 0.1
m
g/ml
Re595 LPS in A
1
medium for 72 h before the cells were labeled with 10
m
g/ml of biotinylated (B)htr-9 or Butr-4 and 0.2
m
g/ml streptavidin-
phycoerythrin (Becton Dickinson, Mountain View, CA) as described
previously (38). Binding of FITC-LPS was estimated by adding 10
m
g/ml
FITC-labeled LPS from S. minnesota 6261 in PBS with 2.5% A
1
serum
(PBS-A
1
buffer) to 1 310
6
SW480/
b
-gal cells for 40 min at 04 °C.
Background fluorescence was estimated by adding 6.2
m
g/ml of FITC-
goat anti-murine Ig (Becton Dikinson). After washing three times
in PBS-A
1
buffer, cells were analyzed in a FACScan flow cytometer
(Becton Dickinson).
RESULTS
LPS Activates the CMV Promoter-enhancer in the Colon Car-
cinoma Cell Line SW480Transcriptional activation of the
CMV promoter-enhancer was measured in a reporter gene
assay in SW480 cells stably transfected with a plasmid con-
taining
b
-gal under control of the CMV promoter-enhancer
(33). Treatment of SW480/
b
-gal cells with LPS resulted in
strong induction of
b
-gal activity (Fig. 1) which shows that LPS
can activate intracellular signals leading to activation of the
CMV promoter-enhancer in SW480 cells. S.minnesota LPS
was found to be several orders of magnitude more potent than
LPS from Escherichia coli or from Pseudomonas. S. minnesota
mutant Re595 LPS gave rise to a higher maximal response
than wild type 6261 LPS, while the lipid A part of S. minnesota
LPS showed very little activity on its own (Fig. 1).
The LPS activity was strongly influenced by human serum
which caused a dose-dependent enhancement of LPS mediated
b
-gal activity (Fig. 2A). The presence of neutralizing anti-CD14
mAb 3C10 completely inhibited LPS-mediated induction of
b
-gal activity (Fig. 2B), and we found that recombinant
(r)CD14 could enhance LPS activity in serum-free medium
(Fig. 2C). Recombinant LPS-binding protein (LBP) had little or
no enhancing effect on the LPS response (Fig. 2C), and LBP did
not further potentiate the rCD14-mediated enhancement of the
LPS response (Fig. 2C). Taken together, the data indicate that
SW480 cells do not express a functional membrane CD14 and
that soluble (s)CD14 in human serum is necessary for the LPS
response. The LPS effect was not mediated by TNF or LT-
a
as
neutralizing antibodies against TNF or LT-
a
did not inhibit the
LPS response (data not shown).
LPS Induces Activation of Transcription Factor NF
k
Bata
Significantly Slower Rate than TNF—Nuclear extracts from
SW480 cells stimulated with LPS or TNF were analyzed for
trancription factor NF
k
B. LPS was found to induce activation
of NF
k
B in a dose-dependent manner, and S. minnesota Re595
mutant LPS was more potent than wild type (6261) LPS (Fig.
3). The response maximum of LPS was markedly lower than
that of TNF.
The finding that LPS and TNF both induce activation of
FIG.3.Activation of NF
k
B in SW480/
b
-gal cells by S. minnesota
6261 LPS, Re595 LPS, and TNF. A,NF
k
B band shift analysis of
nuclear extracts from cells stimulated for 2 h with increasing amounts
of TNF (lanes 1–6), Re595 LPS (lanes 7–12), or 6261 LPS (lanes 13–18),
in A
1
medium. The two bands marked with arrowheads represent
nuclear proteins binding specifically to the NF
k
B consensus sequence,
as identified previously (20). The faster migrating complex () is only
weakly up-regulated and mainly contains NF
k
B p50 as judged by
supershift analysis.
3
The slower migrating complex () contains both
NF
k
B p50 and p65
3
and is strongly up-regulated. B, OCT band shift
analysis of the same extracts as in A.C, quantitation of relative radio-
activity in the slower migrating, strongly up-regulated p50/p65 NF
k
B
complex () from the band shift analysis shown in A, by PhosphoImager
measurements (mean 6S.D. of triplicate measurements of the same
band shifts). Similar results were obtained by analysis of two other
series of nuclear extracts from cells stimulated as above.
Comparison between LPS and TNF Signal Transduction25420
by guest on November 13, 2016http://www.jbc.org/Downloaded from
transcription factor NF
k
B and activate the CMV promoter-
enhancer in SW480 cells, raises the question whether these
agents mediate their effects through similar intracellular
mechanisms. In order to compare the LPS and TNF responses,
we first analyzed the kinetics of NF
k
B activation for each of the
stimuli. It was found that LPS activated NF
k
B at a signifi-
cantly slower rate than TNF (Fig. 4, Aand B). Activation of
NF
k
B by TNF was clearly detectable at 10 min and reached a
plateau level after approximately 45 min. However, LPS-in-
duced NF
k
B activation was only detected after 60 min and
further increased during incubations of up to 120 min (Fig. 4, A
and B). Stimulation with LPS for more than 2 h did not signif-
icantly increase the amounts of activated NF
k
B (data not
shown). In the reporter gene assay, LPS required at least6hto
yield maximal response, while the TNF response reached pla-
teau levels after4hofincubation (Fig. 4C), indicating that LPS
also activates the CMV promoter-enhancer at a slower rate
than TNF. These results suggest that LPS activates NF
k
B and
CMV promoter-enhancer via mechanisms that are different
from the TNF-activated mechanisms.
Pretreatment with TNF Inhibits LPS-induced Activation of
NF
k
B but LPS Pretreatment Does Not Inhibit TNFR p55 or
TNFR p75 Responses—LPS- and TNF-induced NF
k
B activa-
tion was further compared in a series of experiments where
SW480 cells were pretreated for 72 h with either LPS, TNF, or
agonistic antibodies against TNFR p55 or TNFR p75, followed
by stimulation with LPS, TNF, or agonistic TNFR antibodies.
Pretreatment of the cells for 72 h with TNF only marginally
influenced the expression of TNFR p55 and TNFR p75 recep-
tors (Fig. 5A). Furthermore, pretreatment of the cells with TNF
or LPS did not affect the binding of FITC-LPS from S. minne-
sota (Fig. 5B).
Band shift analysis of nuclear extracts from pretreated cells
stimulated with LPS, TNF, or with agonistic anti-TNFR anti-
bodies are shown in Fig. 6. The data demonstrate that the LPS
response is inhibited by pretreatment with either LPS or with
TNF or agonistic TNFR antibodies (Fig. 6, Aand B), while the
TNF response is only inhibited by pretreatment with TNF or
TNFR antibodies and not by LPS pretreatment (Fig. 6, Cand
D). Activation of NF
k
B by TNFR p55 mAb htr-9 was inhibited
by pretreatment with htr-9 or TNF (Fig. 6, Eand F), while the
TNFR p75-mediated response was mainly inhibited by p75 AS
and TNF pretreatment (Fig. 6, Gand H). Taken together, the
results suggest that pretreatment with a given stimulus leads
to depletion or reduction of active intracellular components
involved in the signal transduction pathway induced by that
stimulant. In addition, pretreatment with agents like htr-9 or
TNF may also reduce the level of active components involved in
signal transduction by other agents like LPS.
In all experiments performed, inhibition of the LPS response
by pretreatment with TNFR p55 mAb htr-9 was of a similar
magnitude as inhibition by LPS pretreatment (Fig. 6, Aand B),
indicating that activation of NF
k
B by LPS is dependent on
cellular components which are reduced or inactivated by long
term stimulation of TNFR p55. Pretreatment with TNFR p75
antiserum led to a weak but consistent inhibition of the LPS
response (Fig. 6, Aand B) and thus, components activated via
TNFR p75 may also be involved in LPS signal transduction.
However, neither TNFR p55- nor TNFR p75-mediated activa-
tion of NF
k
B was inhibited by pretreatment with LPS (Fig. 6,
E-H), suggesting that long term stimulation with LPS does not
lead to a reduction or inactivation of components involved in
either of these pathways. In contrast to the lack of inhibition of
the TNFR p75 response by LPS, pretreatment with TNFR p55
mAb htr-9 caused a low but reproducible inhibition of the
TNFR p75 response. This inhibition, however, was markedly
FIG.4.Kinetics of Re595 LPS- and TNF-mediated activation of NF
k
B and CMV promoter-enhancer in SW480/
b
-gal cells. A, band
shift analysis of nuclear extracts from cells treated for the indicated time points with either Re595 LPS (1
m
g/ml) or with TNF (10 ng/ml) in A
1
medium. The two specific NF
k
B complexes are indicated as in Fig. 3. B, PhosphoImager quantitation of radioactivity in the slower migrating
p50/p65 NF
k
B complex (mean 6S.D. of triplicate measurements of the same band shifts). Similar results were obtained by analysis of two other
series of nuclear extracts from cells stimulated as above. C, induction of
b
-galactosidase activity in cells treated for the indicated time points with
either Re595 LPS (0.1
m
g/ml), TNF (10 ng/ml), or A
1
medium. Results (mean values of duplicates) of a representative experiment are given.
FIG.5.Flowcytometric analysis of TNFR mAb and FITC-LPS
binding to pretreated SW480/
b
-gal cells. Cells were pretreated for
72 h with TNF (1 ng/ml) or LPS (0, 1
m
g/ml) followed by labeling of the
cells with p55/p75 mAb (A) or FITC-LPS (B) as described under
“Materials and Methods.”
Comparison between LPS and TNF Signal Transduction 25421
by guest on November 13, 2016http://www.jbc.org/Downloaded from
lower than the inhibition of the TNFR p75 response caused by
p75 AS pretreatment (Fig. 6, Gand H). The TNFR p55-medi-
ated response was not inhibited by pretreatment with p75 AS
in any of the experiments. Thus, it seems that TNFR p55-
induced activation of NF
k
B does not depend on components
activated via TNFR p75.
Pretreatment with TNFR p55 Agonistic Antibodies Can Also
Suppress LPS-induced Transcriptional Activation of the CMV
Promoter-enhancer—LPS- and TNFR-mediated activation of
the CMV promoter-enhancer was compared by subjecting the
cells to similar protocols of pretreatment and stimulation as
described above. It was found that LPS-induced
b
-gal activity
was markedly reduced after pretreatment with LPS or with the
TNFR p55 mAb htr-9, while pretreatment with p75 AS had less
inhibitory effect (Fig. 7A). The TNFR p55-mediated activity
was almost completely abolished after pretreatment with
TNFR p55 mAb htr-9 and very weakly inhibited by pretreat-
ment with p75 AS, while pretreatment with LPS showed no
inhibitory effect (Fig. 7B). Thus, pretreatment of the cells af-
fected the subsequent activation of both the CMV promoter-
enhancer and NF
k
B in a similar manner.
DISCUSSION
The present paper demonstrates that LPS can induce acti-
vation of transcription factor NF
k
B, as well as activation of the
CMV promoter-enhancer in the human adenocarcinoma cells
SW480. These cells do not express a functional membrane
CD14 because addition of LBP did not enhance the LPS effect
under serum free conditions. Such lack of enhancement is a
typical phenomenon in cells which do not express a functional
membrane CD14 (39, 40). Other CD14 negative cells where
LPS has been found to mediate activation of NF
k
B include the
murine pre-B-cell line 70Z/3 (10) and endothelial cells (11). The
LPS response in SW480 cells was strongly dependent on hu-
FIG.6.Activation of NF
k
B in pretreated SW480/
b
-gal cells by Re595 LPS, TNF, or agonistic TNFR antibodies. Cells were pretreated
of for 72 h with either A
1
medium alone, Re595 LPS (0.1
m
g/ml), TNF (1 ng/ml), TNFR p55 mAb htr-9 (10
m
g/ml) (35), or with TNFR p75 antiserum
p75 AS (dil. 1/100) (20), and band shift analysis performed on nuclear extracts from cells stimulated for 2 h with different doses of Re595 LPS (A),
TNF (C), htr-9 (E), or with p75 AS (G), followed by PhosphoImager quantitation of relative radioactivity in the slower migrating, p50/p65
containing complex B,D,F, and H(mean 6S.D. of triplicate measurements of the same band shifts). Similar results were obtained by analysis
of three other series of nuclear extracts from cells pretreated and stimulated as above.
Comparison between LPS and TNF Signal Transduction25422
by guest on November 13, 2016http://www.jbc.org/Downloaded from
man serum and could be completely inhibited by neutralizing
antibodies against CD14. Recombinant soluble (s)CD14 could
only partly compensate for the human serum enhancing effect
since the activity of LPS in the presence of rCD14 was mark-
edly lower than LPS activity in the presence of 20% human
serum. Thus, sCD14 is necessary but not sufficient for LPS
activity, and other serum factors in addition to sCD14 are
necessary for maximal LPS response in SW480 cells.
LPS stimulates activation of NF
k
B at a markedly slower rate
than TNF, indicating that the LPS and TNF signal transduc-
tion pathways are not identical. As shown earlier, the kinetics
of NF
k
B activation by agonistic antibodies against the TNFR
p55 is identical to the TNF kinetics with maximal levels
reached after ;45 min, while stimulation of TNFR p75 results
in maximum activation after ;60 min (20). Thus, in order to
reach maximum NF
k
B activation, stimulation with LPS has to
be continued for a significantly longer period of time than
stimulation of any of the TNF receptors, indicating that the
LPS signaling mechanism differs from the mechanisms em-
ployed by the two TNF receptors. Supershift analysis showed
that stimulation of SW480 cells with LPS or with agonistic
TNFR p55 or p75 antibodies resulted in activation of an iden-
tical pattern of NF
k
B hetero- and homodimers.
3
Thus, SW480
is a cell system where the different pathways employed by LPS
and the two TNF receptors can be compared, and where the
question can be asked as to whether these pathways are inde-
pendent or overlapping.
Comparison of LPS and TNF signal transduction pathways
was performed by analyzing activation of NF
k
B and the CMV
promoter-enhancer in cells pretreated with LPS, TNF, or with
agonistic antibodies against TNFRs. We found that pretreat-
ment of the cells did not result in down-regulation of TNF
receptors or reduction in the binding of LPS. Thus, it is likely
that the observed inhibition of activation of NF
k
B and CMV
promoter-enhancer is due to intracellular effects of the pre-
treatment analogous to depletion of protein kinase C by long
term treatment with the phorbol ester PMA. Such treatment
renders cells unresponsive to subsequent activation of NF
k
Bby
PMA, while the TNF response remains unaffected, indicating
3
A. Lægreid and L. Thommesen, unpublished observations.
FIG.6—continued
Comparison between LPS and TNF Signal Transduction 25423
by guest on November 13, 2016http://www.jbc.org/Downloaded from
that TNF does not depend on PMA-responsive protein kinase C
for activation of NF
k
B (41, 42).
The results from the pretreatment experiments are in agree-
ment with our previous results which indicated that TNFR p75
mediates NF
k
B activation through a different signal transduc-
tion mechanism than TNFR p55 (20). Thus, the TNFR p75-
mediated response is maximally inhibited by pretreatment
with p75 AS while pretreatment with TNFR p55 mAb htr-9 led
to a markedly lower reduction of the TNFR p75 response.
Furthermore, lack of inhibition of the TNFR p75 response by
LPS pretreatment suggests that the TNFR p75-activated sig-
naling mechanism is independent of components activated by
LPS. Thus, the TNFR p75 pathway leading to NF
k
B activation
is different from the LPS, as well as from the TNFR p55
pathway, although it appears to include intracellular compo-
nents which are depleted or inactivated by long term stimula-
tion of TNFR p55.
TNFR p55, which mediates rapid NF
k
B activation, appar-
ently employs a signaling mechanism which is independent of
intracellular components activated by LPS or TNFR p75,
since pretreatment with LPS or TNFR p75 AS did not inhibit
the TNFR p55 response. On the other hand, the LPS response
seems to be mediated by a pathway which is partly overlap-
ping with the TNFR p55 pathway, since pretreatment with
TNFR p55 mAb htr-9 inhibited the LPS response to a similar
extent as the LPS pretreatment. The observation that TNFR
p55 and LPS signaling pathways may be partly overlapping
suggests that TNFR p55 employs more than one pathway
leading to activation of NF
k
B in SW480 cells. Thus, our
results suggest that TNFR p55 may activate one pathway
which mediates rapid activation of NF
k
B and is independent
of intracellular components activated by LPS or p75 AS, and
another pathway which overlaps with the LPS signal trans-
duction pathway.
The stage at which the LPS and TNFR p55 signaling path-
ways overlap may involve ceramide, a lipid messenger which
participates in the activation of NF
k
B in several cell lines
including Jurkat (23), HL-60 (43), and SW480 (42). TNFR
p55-mediated activation of NF
k
B in Jurkat cells as well as
70Z/3 cells has been found to proceed by ceramide generated by
an acidic sphingomyelinase (23, 44), while TNFR p55-mediated
activation of NF
k
B in HL-60 cells is reported to involve a
97-kDa ceramide-activated protein kinase which is activated
via ceramide generated by a neutral sphingomyelinase (43, 45,
46). Recently, LPS was found to stimulate ceramide-activated
protein kinase in HL-60 cells directly, in the absence of detect-
able sphingomyelinase activity (47). A possible reason for this
LPS activity may be structural similarities between LPS and
ceramide (47). Thus, ceramide-activated protein kinase may be
an intracellular component putatively involved in both LPS-
and TNFR p55-mediated NF
k
B activation in SW480 cells.
Release of LPS during Gram-negative infections may induce
high levels of circulating TNF which can lead to shock and
death (48). Our finding that TNF pretreatment inhibits LPS-
induced NF
k
B activation may have important clinical implica-
tions as release of low TNF levels during Gram-negative infec-
tions could render cells resistant to subsequent LPS
stimulation. This is supported by in vivo data showing that
pretreatment of mice with TNF or IL-1 induces partial toler-
ance to LPS (49). Thus, release of low TNF levels during Gram-
negative infections may have an important function in limiting
harmful effects of LPS in vivo.
Acknowledgments—We thank Siv Moen, Wenche Rikardsen, Liv
Ryan, and Mari Sørensen for excellent technical assistance.
REFERENCES
1. Beutler, B., Milsark, I. W., and Cerami, A. C. (1985) Science 229, 869–871
2. Morrison, D. C., and Ryan, J. L. (1987) Annu. Rev. Med. 38, 417–432
3. Ding, A. H., Nathan, C. F., and Stuehr, D. J. (1988) J. Immunol. 141,
2407–2412
4. Wright, S. D., Ramos, R. A., Tobias, P. S., Ulevitch, R. J., and Mathison, J. C.
(1990) Science 249, 1431–1433
5. Couturier, C., Jahns, G., Kazatchkine, M. D., and Haeffner Cavaillon, N.
FIG.7.Induction of
b
-galactosidase activity by Re595 LPS or
by agonistic TNFR p55 mAb htr-9 in pretreated SW480/
b
-gal
cells. Cells were pretreated for 72 h with either A
1
medium alone,
Re595 LPS (0.1
m
g/ml), htr-9 (1
m
g/ml), or with TNFR p75 antiserum
p75 AS (dilution 1/200) followed by stimulation for 4 h with Re595 LPS
(A) or htr-9 (B). Results (mean values of duplicates) of a representative
experiment are given.
Comparison between LPS and TNF Signal Transduction25424
by guest on November 13, 2016http://www.jbc.org/Downloaded from
(1992) Eur. J. Immunol. 22, 1461–1466
6. Lynn, W. A., Liu, Y., and Golenbock, D. T. (1993) Infect. Immun. 61,
4452–4461
7. Pedron, T., Girard, R., Turco, S. J., and Chaby, R. (1994) J. Biol. Chem. 269,
2426–2432
8. Lynn, W. A., and Golenbock, D. T. (1992) Immunol. Today 13, 271–276
9. Frey, E. A., Miller, D. S., Jahr, T. G., Sundan, A., Bazil, V., Espevik, T., Finlay,
B. B., and Wright, S. D. (1992) J. Exp. Med. 176, 1665–1671
10. Sen, R., and Baltimore, D. (1986) Cell 47, 921–928
11. Read, M. A., Cordle, S. R., Veach, R. A., Carlisle, C. D., and Hawiger, J. (1993)
Proc. Natl. Acad. Sci. U. S. A. 90, 9887–9891
12. Godambe, S. A., Chaplin, D. D., Takova, T., and Bellone, C. J. (1994)
J. Immunol. 153, 143–152
13. Zhang, Y., Broser, M., and Rom, W. N. (1994) Proc. Natl. Acad. Sci. U. S. A. 91,
2225–2229
14. Kronke, M., Schutze, S., Scheurich, P., and Pfizenmaier, K. (1992) Immunol.
Ser. 56, 189–216
15. Baeuerle, P. A., and Henkel, T. (1994) Annu. Rev. Immunol. 12, 141–179
16. Beg, A. A., Finco, T. S., Nantermet, P. V., and Baldwin, A. S. (1993) Mol. Cell.
Biol. 13, 3301–3310
17. Henkel, T., Machleidt, T., Alkalay, I., Kronke, M., Ben Neriah, Y., and
Baeuerle, P. A. (1993) Nature 365, 182–185
18. Palombella, V. J., Rando, O. J., Goldberg, A. L., and Maniatis, T. (1994) Cell
78, 773–785
19. Thompson, J. E., Phillips, R. J., Erdjument-Bromage, H., Tempst, P., and
Ghosh, S. (1995) Cell 80, 573–582
20. Laegreid, A., Medvedev, A., Nonstad, U., Bombara, M. P., Ranges, G., Sundan,
A., and Espevik, T. (1994) J. Biol. Chem. 269, 7785–7791
21. Hohmann, H. P., Brockhaus, M., Baeuerle, P. A., Remy, R., Kolbeck, R., and
van Loon, A. P. (1990) J. Biol. Chem. 265, 22409–22417
22. Kruppa, G., Thoma, B., Machleidt, T., Wiegmann, K., and Kronke, M. (1992)
J. Immunol. 148, 3152–3157
23. Schutze, S., Potthoff, K., Machleidt, T., Berkovic, D., Wiegmann, K., and
Kronke, M. (1992) Cell 71, 765–776
24. Wiegmann, K., Schutze, S., Machleidt, T., Witte, D., and Kronke, M. (1994)
Cell 78, 1005–1015
25. Darnay, B. G., Reddy, S. A., and Aggarwal, B. B. (1994) J. Biol. Chem. 269,
19687–19690
26. VanArsdale, T. L., and Ware, C. F. (1994) J. Immunol. 153, 3043–3050
27. Darnay, B. G., Reddy, S. A., and Aggarwal, B. B. (1994) J. Biol. Chem. 269,
20299–20304
28. Rothe, M., Wong, S. C., Henzel, W. J., and Goeddel, D. V. (1994) Cell 78,
681–692
29. Geng, Y., Zhang, B., and Lotz, M. (1993) J. Immunol. 151, 6692–6700
30. Reddy, S. A., Chaturvedi, M. M., Darnay, B. G., Chan, H., Higuchi, M., and
Aggarwal, B. B. (1994) J. Biol. Chem. 269, 25369–25372
31. Schreck, R., Meier, B., Mannel, D. N., Droge, W., and Baeuerle, P. A. (1992)
J. Exp. Med. 175, 1181–1194
32. Ziegler Heitbrock, H. W., Sternsdorf, T., Liese, J., Belohradsky, B., Weber, C.,
Wedel, A., Schreck, R., Bauerle, P., and Strobel, M. (1993) J. Immunol. 151,
6986–6993
33. Galloway, C. J., Madanat, M. S., Sarr, T., Espevik, T., Dumas, M. L., Mitra, G.,
and Ranges, G. E. (1992) Eur. J. Immunol. 22, 3045–3048
34. Hailman, E., Lichenstein, H. S., Wurfel, M. M., Miller, D. S., Johnson, D. A.,
Kelley, M., Busse, L. A., Zukowski, M. M., and Wright, S. D. (1994) J. Exp.
Med. 179, 269–277
35. Brockhaus, M., Schoenfeld, H. J., Schlaeger, E. J., Hunziker, W., Lesslauer,
W., and Loetscher, H. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 3127–3131
36. Updyke, T. V., and Nicolson, G. L. (1984) J. Immunol. Methods 73, 83–95
37. Sturm, R. A., Das, G., and Herr, W. (1988) Genes & Dev. 2, 1582–1599
38. Espevik, T., Brockhaus, M., Loetscher, H., Nonstad, U., and Shalaby, R. (1990)
J. Exp. Med. 171, 415–426
39. Lee, J. D., Kato, K., Tobias, P. S., Kirkland, T. N., and Ulevitch, R. J. (1992)
J. Exp. Med. 175, 1697–1705
40. Espevik, T., Otterlei, M., Skjak Braek, G., Ryan, L., Wright, S. D., and Sundan,
A. (1993) Eur. J. Immunol. 23, 255–261
41. Meichle, A., Schutze, S., Hensel, G., Brunsing, D., and Kronke, M. (1990)
J. Biol. Chem. 265, 8339–8343
42. Johns, L. D., Sarr, T., and Ranges, G. E. (1994) J. Immunol. 152, 5877–5882
43. Yang, Z., Costanzo, M., Golde, D. W., and Kolesnick, R. N. (1993) J. Biol.
Chem. 268, 20520–20523
44. Machleidt, T., Wiegmann, K., Henkel, T., Schutze, S., Baeuerle, P., and
Kronke, M. (1994) J. Biol. Chem. 269, 13760–13765
45. Mathias, S., Dressler, K. A., and Kolesnick, R. N. (1991) Proc. Natl. Acad. Sci.
U. S. A. 88, 10009–10013
46. Liu, J., Mathias, S., Yang, Z., and Kolesnick, R. N. (1994) J. Biol. Chem. 269,
3047–3052
47. Joseph, C. K., Wright, S. D., Bornmann, W. G., Randolph, J. T., Kumar, E. R.,
Bittman, R., Liu, J., and Kolesnick, R. N. (1994) J. Biol. Chem. 269,
17606–17610
48. Waage, A., Halstensen, A., and Espevik, T. (1987) Lancet 1, 355–357
49. Erroi, A., Fantuzzi, G., Mengozzi, M., Sironi, M., Orencole, S. F., Clark, B. D.,
Dinarello, C. A., Isetta, A., Gnocchi, P., Giovarelli, M., and et al. (1993)
Infect. Immun. 61, 4356–4359
Comparison between LPS and TNF Signal Transduction 25425
by guest on November 13, 2016http://www.jbc.org/Downloaded from
Astrid Lægreid, Liv Thommesen, Tove Gullstein Jahr, Anders Sundan and Terje Espevik
Adenocarcinoma Cell Line Mainly through the TNF p55 Receptor
Tumor Necrosis Factor Induces Lipopolysaccharide Tolerance in a Human
doi: 10.1074/jbc.270.43.25418
1995, 270:25418-25425.J. Biol. Chem.
http://www.jbc.org/content/270/43/25418Access the most updated version of this article at
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/270/43/25418.full.html#ref-list-1
This article cites 49 references, 32 of which can be accessed free at
by guest on November 13, 2016http://www.jbc.org/Downloaded from
... TNF tolerance can be induced by a pretreatment of monocytes and macrophages (or other eligible cells) with both low and high doses of TNF [3,4,26]. Typically, low-dose preexposure/incubation was performed using up to 10 μg/kg (mostly human) TNF in animal experiments or up to 20 ng/ml in cell culture experiments [3,27], while high doses included up to 100 μg/kg in animal studies [2]. It also has to be taken into account that human TNF can induce tolerance in animal experiments using higher doses due to a reduced cytotoxicity, for example, in comparison to murine TNF which proved to be lethal at significantly lower doses [19]. ...
... Thus, TNF tolerance appears to be mediated via TNFR1 [16,19]. While total amounts of TNFR1 were not significantly affected during long-term treatment in monocytic THP-1 (high-dose TNF) or SW480 epithelial cells (low-dose TNF) [27,36], TNF stimulation led to an internalization of the receptor within 2 h irrespective of the dose used [36]. Application of low-dose TNF during the preincubation phase, however, resulted in a (slow) recurrence of the initial level of TNFR1 within 48 h. ...
... The TNF-tolerant state may further include the refractoriness towards other stimuli. For instance, cells or animals tolerized with TNF are cross-tolerant towards gram-negative bacteria [7,18], LPS [2,6,27], or other bacteria-derived agents such as lipophilic outer membrane vesicles [7]. Vice versa pretreatment with LPS [5,48] or macrophageactivating lipopeptide 2 [49] induced the development of tolerance towards subsequent TNF application in THP-1 cells or mice and rats. ...
Article
Full-text available
Tumor necrosis factor (TNF) tolerance in monocytes and macrophages means that preexposure to TNF reduces the sensitivity in these cells to a subsequent restimulation with this cytokine. Differential effects arise following preincubation with both low and high doses of TNF resulting in absolute as well as induction tolerance affecting specific immunologically relevant gene sets. In this review article, we summarize the relevance of TNF tolerance in vivo and the molecular mechanisms underlying these forms of tolerance including the role of transcription factors and signaling systems. In addition, the characteristics of cross-tolerance between TNF and lipopolysaccharide (LPS) as well as pathophysiological aspects of TNF tolerance are discussed. We conclude that TNF tolerance may represent a protective mechanism involved in the termination of inflammation and preventing excessive or prolonged inflammation. Otherwise, tolerance may also be a trigger of immune paralysis thus contributing to severe inflammatory diseases such as sepsis. An improved understanding of TNF tolerance will presumably facilitate the implementation of diagnostic or therapeutic approaches to more precisely assess and treat inflammation-related diseases.
... Antigen-specific T-cell anergy by oral tolerance induction was demonstrated by the transfer of T cells and B cells from orally tolerized mice into SCID mice [15]. The clonal deletion process occurs by apoptosis of antigen-specific CD4 + T cells [16], which in oral tolerance induction is mediated by signaling via Fas antigen and p55 tumor necrosis factor (TNF) receptor [17,18]. Various regulatory T cells are induced by oral tolerance induction. ...
Article
Full-text available
Food allergy is a serious problem for infants and young children. Induction of antigen-specific oral tolerance is one therapeutic strategy. Enhancement of oral tolerance induction by diet is a promising strategy to prevent food allergy in infants. Thus, in this study, we evaluate the effect of probiotic Lactobacillus gasseri OLL2809 (LG2809) on oral tolerance induction in a mouse model. The degree of oral tolerance induction was evaluated by measuring the proliferation and level of IL-2 production of splenic CD4+ T cells from DO11.10 mice fed ovalbumin (OVA) alone or OVA with LG2809. Oral administration of LG2809 significantly decreased the rate of proliferation and IL-2 production by CD4+ T cells from OVA-fed mice. LG2809 increased a ratio of CD4+ T-cell population, producing high levels of IL-10 and having strong suppressive activity. Moreover, LG2809 increased a ratio of plasmacytoid dendritic cells (pDCs) among the lamina propria (LP) in small intestine. When used as antigen presenting cells to naïve CD4+ T cells from DO11.10 mice, LP cells from BALB/c mice fed LG2809 induced higher IL-10 production and stronger suppressive activity than those from non-treated mice. These results suggest that oral administration of LG2809 increases the population of pDCs in the LP, resulting in the enhancement of oral tolerance induction by increasing the ratio of effector regulatory T cells. LG2809 could, therefore, act as a potent immunomodulator to prevent food allergies by promoting oral tolerance.
... Because TNF tolerance appears more slowly than that of LPS, different mechanisms seem to be responsible for the two phenomena (14). Only a few results from cell culture studies characterizing the molecular basis of TNF tolerance exist to date (9,15,16). At the beginning of this study, it was unclear whether the phenomenon of TNF tolerance exists in primary monocytes as major producers of TNF coordinating innate and adaptive immunity (17). ...
Article
Full-text available
The molecular basis of TNF tolerance is poorly understood. In human monocytes we detected two forms of TNF refractoriness, as follows: absolute tolerance was selective, dose dependently affecting a small group of powerful effector molecules; induction tolerance represented a more general phenomenon. Preincubation with a high TNF dose induces both absolute and induction tolerance, whereas low-dose preincubation predominantly mediates absolute tolerance. In cells preincubated with the high TNF dose, we observed blockade of IκBα phosphorylation/proteolysis and nuclear p65 translocation. More prominent in cells preincubated with the high dose, reduced basal IκBα levels were found, accompanied by increased IκBα degradation, suggesting an increased IκBα turnover. In addition, a nuclear elevation of p50 was detected in tolerant cells, which was more visible following high-dose preincubation. TNF-induced phosphorylation of p65-Ser(536), p38, and c-jun was inhibited, and basal inhibitory p65-Ser(468) phosphorylation was increased in tolerant cells. TNF tolerance induced by the low preincubation dose is mediated by glycogen synthesis kinase-3, whereas high-dose preincubation-mediated tolerance is regulated by A20/glycogen synthesis kinase-3 and protein phosphatase 1-dependent mechanisms. To our knowledge, we present the first genome-wide analysis of TNF tolerance in monocytic cells, which differentially inhibits NF-κB/AP-1-associated signaling and shifts the kinase/phosphatase balance. These forms of refractoriness may provide a cellular paradigm for resolution of inflammation and may be involved in immune paralysis.
... The colonic cell lines display important functional and morphological characteristics of human intestinal epithelial cells [34]. All cell lines employed were confirmed previously to respond to treatment with TNFα [35,36,37]. ...
Article
Butyrate, a short-chain fatty acid, is generated by anaerobic fermentation within the colon. Clinical trials suggest that short-chain fatty acids ameliorate inflammation in ulcerative colitis. Nuclear factor (NF) &#51B, an inducible transcription factor that is activated in inflamed colonic tissue, is sequestered to the cytoplasm by its inhibitory I&#51B proteins. The anti-inflammatory effects of butyrate are associated with an inhibition of NF-&#51B nuclear translocation. To investigate the mechanism of NF-&#51B inhibition we examined the effects of butyrate on I&#51B&#33. Human adenocarcinoma cells (SW480, SW620, and HeLa229) were treated with butyrate for up to 48 h followed by tumor necrosis factor (TNF) ! stimulation. NF-&#51B was detected by immunofluorescence staining with an antibody against its p65 subunit. Levels of I&#51B! and phosphorylated I&#51B! were determined by western blot. Stimulation with TNF! resulted in rapid phosphorylation and degradation of I&#51B! followed by NF-&#51B nuclear translocation. Butyrate pretreatment successfully inhibited NF-&#51B activation. Pretreatment of adenocarcinoma cells with butyrate is associated with inhibition of TNF&#33-mediated phosphorylation and degradation of I&#51B! and effective blocking of NF-&#51B nuclear translocation. The anti-inflammatory effects of butyrate may at least in part be mediated by an inhibition of I&#51B! mediated activation of NF-&#51B.
... Expression of LVV-EAAT2 is driven by the CMV-promoter, which can be strongly activated by TNF-α, via the induction of the transcription factor nuclear factor-κB (NF-κB) (Prosch et al., 1995;Laegreid et al., 1995). Given the above association between ALS pathogenesis, TNF-α, and endogenous EAAT2 gene transcription, it was anticipated that, in contrast to endogenous EAAT2, the LVV-mediated EAAT2 production would not be inhibited by the high levels of TNF-α such known to be present in the ALS affected brain. ...
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) is a paralytic neurodegenerative disorder, characterised by a specific loss of motoneurons. Although the exact pathogenesis is largely enigmatic, it is known that glutamate excitotoxicity plays an important role in motoneuron cell death. Glutamate is one of the major neurotransmitters in the central nervous system. Relative high levels of glutamate can lead to motoneuron death by overstimulation of calcium-permeable receptors, resulting in an excess influx of calcium. This can lead to activation of several death-inducing pathways. Therefore, it is of great significance to keep the extracellular glutamate levels at a non-toxic value. Under physiological conditions the glutamate transporter EAAT2, predominantly expressed by astrocytes, accomplishes the majority of the glutamate uptake from the synaptic cleft. In ALS patients and in ALS animal models, however, it is found that EAAT2 protein and functioning is decreased. The aim of the studies described in this thesis was to protect motoneurons from excitotoxicity by overexpressing the glutamate transporter EAAT2 in a localised, long-term manner via gene therapy, both ex vivo and in vivo gene therapy are exploited for this purpose. In the first part of this thesis we examined the possibilities for ex vivo gene therapy. To this end, HEK-cells were genetically engineered to overexpress EAAT2. This resulted in an increase in EAAT2 protein as well as a profuse increase in glutamate uptake compared to wild-type cells. Furthermore, the engineered cells protected primary motoneurons in culture from an glutamate-induced toxicity. Hence, it might be worthwhile to investigate whether ex vivo gene therapy for EAAT2 might be of therapeutic value in amyotrophic lateral sclerosis. In the second part of this thesis we further examined the possibilities for in vivo gene therapy with the use of lentiviral vectors (LVV) to overexpress EAAT2. For this purpose we first examined the cellular pattern of LVV-mediated gene transduction in mouse spinal cord both in vitro and in vivo. Therefore, organotypic mouse spinal cord cultures were treated with LVV-green fluorescent protein (GFP) in vitro and wild-type mice were intraspinally injected with LVV-GFP at the level of vertebra L1. In both situations GFP expression was found to be expressed predominately by astrocytes, while the neuronal cells were virtually not transduced. Next, we examined whether LVV-EAAT2 could be used as a tool to deliver EAAT2 to primary adult human astrocytes from either cortical or spinal post-mortem brain tissue. Concomitant with a marked increase in EAAT2 mRNA and EAAT2 protein, a significant increase in functional glutamate uptake was observed in LVV-EAAT2 treated astrocytic cultures compared to LVV-GFP treated controls. Finally, we examined the neuroprotective efficacy of intraspinally delivered LVV-EAAT2 on the course of the disease in G93A-hSOD1 ALS mice by using motor performance and body weight as clinical parameters. We found that EAAT2 was effectively transduced in the spinal cord, but that disease onset and survival in the LVV-EAAT2 treated mice did not significantly differ from either LVV-GFP treated or naive control mice. We concluded that, although LVV-EAAT2 did not effect the clinical outcome of ALS mice, as such LVV can be successfully used to deliver therapeutic genes into spinal astrocytes.
... Here blockade has been demonstrated to operate at various levels along the signalling cascade and this includes failure of MyD88 to be recruited to TLR4 [11], up-regulation of IRAK-M [12] or SHIP [13], up-regulation of suppressor of cytokine signalling-1 [14,15] and downregulation of MAP-kinases [16]. A major mechanism of tolerance that can be demonstrated in macrophages but also in T cells and tumor cells is the up-regulation of p50 mRNA and increased translocation of p50-homodimers into the nucleus [1,1718192,202122. More recently the proteolytic degradation of IRAK-1 has been shown to represent another major mech- anism [4,3,232425. ...
Article
Full-text available
While dendritic cells (DCs) can induce tolerance in T cells, little is known about tolerance induction in DCs themselves. We have analysed tolerance induced in human in-vitro generated DCs by repeated stimulation with ligands for TLR4 and TLR2. DCs stimulated with the TLR4 ligand LPS did show a rapid and pronounced expression of TNF mRNA and protein. When DCs were pre-cultured for 2 days with 5 ng LPS/ml then the subsequent response to stimulation with a high dose of LPS (500 ng/ml) was strongly reduced for both TNF mRNA and protein. At the promoter level there was a reduced transactivation by the -1173 bp TNF promoter and by a construct with a tetrameric NF-kappaB motif. Within the signalling cascade leading to NF-kappaB activation we found an ablation of the IRAK-1 adaptor protein in LPS-tolerant DCs. Pre-culture of DCs with the TLR2 ligand Pam3Cys also led to tolerance with respect to TNF gene expression and IRAK-1 protein was ablated in such tolerant cells as well, while IRAK-4 protein levels were unchanged. These data show that TLR-ligands can render DCs tolerant with respect to TNF gene expression by a mechanism that likely involves blockade of signal transduction at the level of IRAK-1.
Article
Interfering with the endotoxin-mediated cytokine cascade is thought to be a promising approach to prevent septic complications in gram-negative infections. The synthetic lipid A analog SDZ MRL 953 has been shown to be protective against endotoxic shock and bacterial infection in preclinical in vivo models. As part of a trial of unspecific immunostimulation in cancer patients, we conducted a double-blind, randomized, vehicle-controlled phase I trial of SDZ MRL 953 to investigate, first, its biologic effects and safety of administration in humans and, second, its influence on reactions to a subsequent challenge of endotoxin (Salmonella abortus equi). Twenty patients were treated intravenously with escalating doses of SDZ MRL 953 or vehicle control, followed by an intravenous application of endotoxin (2 ng/kg of body weight [BW]). Administration of SDZ MRL 953 was safe and well-tolerated. SDZ MRL 953 itself increased granulocyte counts and serum levels of granulocyte colony-stimulating factor (G-CSF ) and interleukin-6 (IL-6), but not of the proinflammatory cytokines tumor necrosis factor-α (TNF-α), IL-1β, and IL-8. Compared with vehicle control, pretreatment with SDZ MRL 953 markedly reduced the release of TNF-α, IL-1β, IL-8, IL-6, and G-CSF, but augmented the increase in granulocyte counts to endotoxin. Induction of tolerance to the endotoxin-mediated cascade of proinflammatory cytokines by pretreatment with SDZ MRL 953 in patients at risk may help to prevent complications of gram-negative sepsis.
Article
The anti-inflammatory effect of piperine has been largely investigated in macrophages, but its activity on epithelial cells in inflammatory settings is unclear. The present study aimed to investigate the effect of piperine on the expression of inflammatory cytokines in lipopolysaccharide (LPS)-stimulated human epithelial-like SW480 and HT-29 cells. Our data showed that although piperine inhibited the proliferation of SW480 and HT-29 cells in a dose-dependent manner, it had low cytotoxicity on these cell lines with 50 % inhibiting concentration (IC50) values greater than 100 μM. As epithelial-like cells, SW480 and HT-29 cells secreted high levels of the chemokine CXCL8 upon LPS stimulation. Importantly, piperine dose-dependently suppressed LPS-induced secretion of CXCL8 and the expression of CXCL8 messenger RNA (mRNA). Although piperine failed to affect the critical inflammatory nuclear factor-κB pathway, it attenuated the c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signaling. Consistent with previous reports, p38 signaling seemed to play a more pronounced role on the CXCL8 expression than JNK signaling since inhibition of p38, instead of JNK, greatly suppressed LPS-induced CXCL8 expression. Collectively, our results indicated that piperine could attenuate the inflammatory response in epithelial cells via downregulating the MAPK signaling and thus the expression of CXCL8, suggesting its potential application in anti-inflammation therapy.
Article
Full-text available
There is some evidence that the potent cytokine tumor necrosis factor (TNF) is able to induce tolerance after repeated stimulation of cells. To investigate the molecular mechanisms mediating this phenomenon, the expression of interleukin-8 (IL-8), which is regulated by transcription factors NF-κB and C/EBPβ, was monitored under TNF tolerance conditions. Pretreatment of monocytic cells for 72 h with low TNF doses inhibited TNF-induced (restimulation with a high dose) IL-8 promoter-dependent transcription as well as IL-8 production. Under these conditions neither activation of NF-κB nor IκB proteolysis was affected after TNF re-stimulation, albeit a slightly reduced IκB-α level was found in the TNF pretreated but not re-stimulated sample. Remarkably, in tolerant cells an increased binding of C/EBPβ to its IL-8 promoter-specific DNA motif as well as an elevated association of C/EBPβ protein with p65-containing NF-κB complexes was observed. Finally, overexpression of C/EBPβ, but not p65 or Oct-1, markedly prevented TNF-induced IL-8 promoter-dependent transcription. Taken together, these data indicate that the expression of IL-8 is inhibited at the transcriptional level in TNF-tolerant cells and C/EBPβ is involved under these conditions in mediating the negative-regulatory effects, a mechanism that may play a role in inflammatory processes such as sepsis.
Article
Cytokines are recognized to play an important role in acute stroke. Tumor necrosis factor-alpha (TNF) is one of the pro-inflammatory cytokines and is expressed in ischemic brain. We hypothesized that TNF might play a role in the regulation of tolerance to ischemia when administered prior to the ischemic episode. We studied the effects of pretreatment of TNF administered intravenously, intraperitoneally, or intracisternally in mice that were subjected to middle cerebral artery occlusion (MCAO) 48 h later. MCAO was performed in BALB/C mice by direct cauterization of distal MCA, which resulted in pure cortical infarction. A significant reduction in infarct size was noted in mice pretreated by TNF at the dose of 0.5 microgram/mouse (p < 0.01) intracisternally. At the doses used in this study, administration of TNF by intravenous or intraperitoneal routes was not effective. Immunohistochemical analysis of brains subjected to 24 h of MCAO revealed a significant decrease in CD11b immunoreactivity after TNF pretreatment compared with control MCAO. Preconditioning with TNF affects infarct size in a time- and dose-dependent manner. TNF induces significant protection against ischemic brain injury and is likely to be involved in the signaling pathways that regulate ischemic tolerance.
Article
Full-text available
Dithiocarbamates and iron chelators were recently considered for the treatment of AIDS and neurodegenerative diseases. In this study, we show that dithiocarbamates and metal chelators can potently block the activation of nuclear factor kappa B (NF-kappa B), a transcription factor involved in human immunodeficiency virus type 1 (HIV-1) expression, signaling, and immediate early gene activation during inflammatory processes. Using cell cultures, the pyrrolidine derivative of dithiocarbamate (PDTC) was investigated in detail. Micromolar amounts of PDTC reversibly suppressed the release of the inhibitory subunit I kappa B from the latent cytoplasmic form of NF-kappa B in cells treated with phorbol ester, interleukin 1, and tumor necrosis factor alpha. Other DNA binding activities and the induction of AP-1 by phorbol ester were not affected. The antioxidant PDTC also blocked the activation of NF-kappa B by bacterial lipopolysaccharide (LPS), suggesting a role of oxygen radicals in the intracellular signaling of LPS. This idea was supported by demonstrating that treatment of pre-B and B cells with LPS induced the production of O2- and H2O2. PDTC prevented specifically the kappa B-dependent transactivation of reporter genes under the control of the HIV-1 long terminal repeat and simian virus 40 enhancer. The results from this study lend further support to the idea that oxygen radicals play an important role in the activation of NF-kappa B and HIV-1.
Article
Full-text available
Three different antibodies against a human TNF receptor (htr-1, htr-5, and htr-9) have been examined for their binding pattern to U937 cells and ability to mimic TNF-alpha activity in U937 cells, Fs4 fibroblasts, and human endothelial cells. Flow cytometric analysis revealed that htr-5 and htr-9 bound specifically to a TNF receptor on U937 cells that could be blocked by pretreatment with rTNF-alpha. Pretreatment of U937 cells with rTNF-beta blocked the binding of htr-9, but to a lesser extent htr-5 binding. Pretreatment with htr-5 inhibited the binding of htr-9 to U937 cells while pretreatment with htr-9 did not inhibit htr-5 binding. These results indicate that htr-5 and htr-9 recognize distinct but overlapping epitopes of a human TNF receptor on U937 cells and that htr-5 may be close to a TNF-alpha-specific domain of the binding site. Pretreatment with htr-5 or htr-9 only minimally reduced binding of BrTNF-alpha to U937 cells; however, these antibodies were much more effective in inhibiting BrTNF-alpha binding to HL-60 cells. Furthermore, it was found that htr-1 and htr-9, but not htr-5, had TNF-alpha activity on U937 cells, Fs4 fibroblasts, and endothelial cells and that the TNF-alpha activity induced by htr-9 was completely inhibited by htr-5. However, the cytotoxic activity of TNF-alpha was only partially inhibited by htr-5 on U937 cells while htr-5 had no effect on TNF-alpha activity on Fs4 cells. The data suggest that a common epitope is involved in inducing TNF-alpha activity in three different cell systems.
Article
Full-text available
CD14 is a 55-kD protein found both as a glycosylphosphatidyl inositol-linked protein on the surface of mononuclear phagocytes and as a soluble protein in the blood. CD14 on the cell membrane (mCD14) has been shown to serve as a receptor for complexes of lipopolysaccharide (LPS) with LPS binding protein, but a function for soluble CD14 (sCD14) has not been described. Here we show that sCD14 enables responses to LPS by cells that do not express CD14. We have examined induction of endothelial-leukocyte adhesion molecule 1 expression by human umbilical vein endothelial cells, interleukin 6 secretion by U373 astrocytoma cells, and cytotoxicity of bovine endothelial cells. None of these cell types express mCD14, yet all respond to LPS in a serum-dependent fashion, and all responses are completely blocked by anti-CD14 antibodies. Immunodepletion of sCD14 from serum prevents responses to LPS, and the responses are restored by addition of sCD14. These studies suggest that a surface anchor is not needed for the function of CD14 and further imply that sCD14 must bind to additional proteins on the cell surface to associate with the cell and transduce a signal. They also indicate that sCD14 may have an important role in potentiating responses to LPS in cells lacking mCD14.
Article
Full-text available
Dithiocarbamates and iron chelators were recently considered for the treatment of AIDS and neurodegenerative diseases. In this study, we show that dithiocarbamates and metal chelators can potently block the activation of nuclear factor kappa B (NF-kappa B), a transcription factor involved in human immunodeficiency virus type 1 (HIV-1) expression, signaling, and immediate early gene activation during inflammatory processes. Using cell cultures, the pyrrolidine derivative of dithiocarbamate (PDTC) was investigated in detail. Micromolar amounts of PDTC reversibly suppressed the release of the inhibitory subunit I kappa B from the latent cytoplasmic form of NF-kappa B in cells treated with phorbol ester, interleukin 1, and tumor necrosis factor alpha. Other DNA binding activities and the induction of AP-1 by phorbol ester were not affected. The antioxidant PDTC also blocked the activation of NF-kappa B by bacterial lipopolysaccharide (LPS), suggesting a role of oxygen radicals in the intracellular signaling of LPS. This idea was supported by demonstrating that treatment of pre-B and B cells with LPS induced the production of O2- and H2O2. PDTC prevented specifically the kappa B-dependent transactivation of reporter genes under the control of the HIV-1 long terminal repeat and simian virus 40 enhancer. The results from this study lend further support to the idea that oxygen radicals play an important role in the activation of NF-kappa B and HIV-1.
Article
The human TNF promoter contains four potential nuclear factor-kappa B (NF-kappa B)-binding sites, with the strongest binding seen for the -605 motif. Nuclear extracts from unstimulated cells of the human monocytic cell line, Mono Mac 6, contain one specific binding protein (complex II), consistent with a constitutive p50 homodimer. Stimulation of Mono Mac 6 cells with LPS will increase complex II and will strongly induce a second specific complex (complex I), which represents the p50/65 heterodimer. Treatment of Mono Mac 6 cells with pyrrolidine-dithiocarbamate (PDTC) at 300 microM will block the LPS-induced complex I almost completely and will reduce complex II to the constitutive level. Binding activity of other nuclear factors that recognize the SP-1 and c/EBP motifs of the human TNF promoter is not affected by such treatment. Northern blot analysis demonstrates that PDTC treatment will strongly reduce LPS-induced TNF transcripts. Secreted TNF protein as detected in the Wehi 164S/ActD bioassay and in a sandwich immunoassay was similarly reduced by PDTC. Kinetic analyses show that after LPS stimulation, NF-kappa B will peak at 1 h, TNF transcript prevalence at 2 h, and TNF protein at 4 h. PDTC did not shift this response to LPS to a later time, but suppressed NF-kappa B mobilization, TNF transcripts, and TNF protein over the entire 8-h observation period. Analysis of freshly isolated, LPS-stimulated blood monocytes showed a similar blockade of NF-kappa B. Furthermore, in these primary cells, induction of TNF transcripts, as determined by Northern blot analysis and by quantitative polymerase chain reaction, was prevented by PDTC as was TNF protein production. These data show that dithiocarbamates can profoundly affect cytokine expression and suggest that NF-kappa B is involved in LPS-induced TNF gene expression in human monocytes.
Article
Bacterial endotoxin (lipopolysaccharide [LPS]) causes fatal shock in humans and experimental animals. The shock is mediated by cytokines released by direct LPS stimulation of cells of monocytic origin (monocyte/macrophage [MO]). Recent studies have supported the concept that the plasma protein, LPS binding protein (LBP), plays an important role in controlling MO responses to LPS. Specifically, evidence has been presented to suggest that CD14, a membrane protein present in MO, serves as a receptor for complexes of LPS and the plasma protein LPS binding protein (LBP). In this function CD14 mediates attachment of LPS-bearing particles opsonized with LBP and appears to play an important role in regulating cytokine production induced by complexes of LPS and LBP. The CD14-, murine pre-B cell line 70Z/3 responds to LPS by synthesis of kappa light chains and consequent expression of surface IgM. To better understand the role of CD14 in controlling cellular responses to LPS, we investigated the effect of transfection of CD14 into 70Z/3 cells on LPS responsiveness. We report here that transfection of human or rabbit CD14 cDNA into 70Z/3 cells results in membrane expression of a glycosyl-phosphatidylinositol-anchored CD14. When LPS is complexed with LBP, CD14-bearing 70Z/3 cells bind more LPS than do the parental or 70Z/3 cells transfected with vector only. Remarkably, the expression of CD14 lowers the amount of LPS required to stimulate surface IgM expression by up to 10,000-fold when LPS dose-response curves in the CD14-, parental and CD14-bearing, transfected 70Z/3 cells are compared. In contrast, the response of CD14-bearing 70Z/3 cells and the parental 70Z/3 cell line (CD14-) to interferon gamma is indistinguishable. LPS stimulation of the parental and CD14-bearing 70Z/3 cells results in activation of NF-kB. These data provide evidence to support the concept that the LPS receptor in cells that constitutively express CD14 may be a multiprotein complex containing CD14 and membrane protein(s) common to a diverse group of LPS-responsive cells.
Article
CD14 is a 55-kD protein found as a glycosylphosphatidylinositol (GPI)-anchored protein on the surface of monocytes, macrophages, and polymorphonuclear leukocytes, and as a soluble protein in the blood. Both forms of CD14 participate in the serum-dependent responses of cells to bacterial lipopolysaccharide (LPS). While CD14 has been described as a receptor for complexes of LPS with LPS-binding protein (LBP), there has been no direct evidence showing whether a ternary complex of LPS, LBP, and CD14 is formed, or whether CD14 binds LPS directly. Using nondenaturing polyacrylamide gel electrophoresis (native PAGE), we show that recombinant soluble CD14 (rsCD14) binds LPS in the absence of LBP or other proteins. Binding of LPS to CD14 is stable and of low stoichiometry (one or two molecules of LPS per rsCD14). Recombinant LBP (rLBP) does not form detectable ternary complexes with rsCD14 and LPS, but it does accelerate the binding of LPS to rsCD14. rLBP facilitates the interaction of LPS with rsCD14 at substoichiometric concentrations, suggesting that LBP functions catalytically, as a lipid transfer protein. Complexes of LPS and rsCD14 formed in the absence of LBP or other serum proteins strongly stimulate integrin function on PMN and expression of E-selectin on endothelial cells, demonstrating that LBP is not necessary for CD14-dependent stimulation of cells. These results suggest that CD14 acts as a soluble and cell surface receptor for LPS, and that LBP may function primarily to accelerate the binding of LPS to CD14.
Article
We have cloned the cDNA encoding IκB-β, one of the two major IκB isoforms in mammalian cells. The recombinant IκB-β protein interacts with equal affinity to p65 and c-Rel and does not exhibit a preference between these Rel proteins. Instead the primary difference between IκB-α and IκB-β is in their response to different inducers of NF-κB activity. One class of inducers causes rapid but transient activation of NF-κB by primarily affecting IκB-α complexes, whereas another class of inducers causes persistent activation of NF-κB by affecting both IκB-α and IκB-β complexes. Therefore, the overall activation of NF-κB consists of two overlapping phases, a transient phase mediated through IκB-α and a persistent phase mediated through IκB-β.
Article
The numerous biologic activities of TNF appear mediated by two types of specific cell surface receptors of 55 to 60 kDa (TR55) and 75 to 80 kDa (TR75) molecular mass, respectively. The role of TR55 in the activation of the nuclear transcription factor kappa B (NF-kappa B) was investigated using an antagonistic, mAb, H398, specific for the human TR55. The human leukemic T cell line, Jurkat, which expresses both types of receptors at comparable levels, was used to test for NF-kappa B activation by electrophoretic mobility shift assays using as a probe an oligonucleotide encompassing the two tandemly arranged kappa B sites of the HIV-1 LTR enhancer. mAb H398 is shown to efficiently block not only TNF- but also lymphotoxin-mediated activation of NF-kappa B. Furthermore mAb H398 also impeded TNF- or lymphotoxin-mediated activation of chloramphenicol acetyl transferase gene expression from the HIV-1-LTR as determined by transient transfection assays. These findings indicate that both, induction of NF-kappa B binding to DNA, and transcriptional activity can be efficiently inhibited by selective blockade of TR55. Finally it is shown, that human TR55 confers NF-kappa B inducibility when expressed in the mouse pre-B cell line 70Z/3, which does not respond to TNF in its parental state. Together, the results of this study indicate that TR55 is both necessary and sufficient for mediating TNF activation of NF-kappa B.
Article
In this paper, we describe a phospholipid transmission pathway mediating tumor necrosis factor (TNF) activation of the nuclear transcription factor kappa B (NF-kappa B). Central to this TNF signaling route is the second messenger-like molecule ceramide, which is generated by sphingomyelin (SM) breakdown catalyzed by a sphingomyelinase (SMase). SMase activation is secondary to the generation of 1,2-diacylglycerol (DAG) produced by a TNF-responsive PC-specific phospholipase C (PC-PLC). The functional coupling of these two C type phospholipases is revealed by D609, a selective inhibitor of PC-PLC. SMase itself, or SMase-inducing regimens such as exogenous PLC or synthetic DAGs, induces NF-kappa B activation at pH 5.0, suggesting the operation of an acidic SMase. A model is proposed in which a TNF-responsive PC-PLC via DAG couples to an acidic SMase, resulting in the generation of ceramide, which eventually triggers rapid induction of nuclear NF-kappa B activity.