ArticlePDF Available

Tartaglia LA, Weber RF, Figari IS, Reynolds C, Palladino Jr MA, Goeddel DVThe two different receptors for tumor necrosis factor mediate distinct cellular responses. Proc Natl Acad Sci USA 88:9292-9296

Authors:

Abstract and Figures

The individual roles of the murine type 1 and type 2 tumor necrosis factor (TNF) receptors (TNF-R1 and TNF-R2) were investigated utilizing (i) the strong species specificity of TNF-R2 for murine TNF compared to human TNF and (ii) agonistic rabbit polyclonal antibodies directed against the individual TNF receptors. Proliferation of mouse thymocytes and the murine cytotoxic T-cell line CT-6 is stimulated by murine TNF but not by human TNF. Consistent with this observation, polyclonal antibodies directed against TNF-R2 induced proliferation in both of these cell types, whereas polyclonal antibodies directed against TNF-R1 had no effect. In contrast, cytotoxicity in murine LM cells (which are sensitive to murine and human TNF) was induced by antibodies against TNF-R1 but not by antibodies against TNF-R2. Also, the steady-state level of manganous superoxide dismutase mRNA in the murine NIH 3T3 cell line was induced by murine TNF, human TNF, and anti-TNF-R1 but not by anti-TNF-R2. These results suggest that TNF-R2 initiates signals for the proliferation of thymocytes and cytotoxic T cells, whereas TNF-R1 initiates signals for cytotoxicity and the induction of the protective activity, manganous superoxide dismutase. The nonredundant signaling observed for the two TNF receptors cannot be explained simply by the differential expression of the two TNF receptors in the various cell types, because LM cells express on their surface higher levels of TNF-R2 than TNF-R1, and LM cells, NIH 3T3 cells, and thymus cells all express mRNA corresponding to both receptor types. It is therefore likely that the two receptors initiate distinct signaling pathways that result in the induction of different cellular responses.
Content may be subject to copyright.
Proc.
Nati.
Acad.
Sci.
USA
Vol.
88,
pp.
9292-92%,
October
1991
Immunology
The
two
different
receptors
for
tumor
necrosis
factor
mediate
distinct
cellular
responses
(species
specificity/antibodies/manganous
superoxide
dismutase/cytotoxicity/proliferation)
Louis
A.
TARTAGLIA*,
RICHARD
F.
WEBER*,
IRENE
S.
FIGARIt,
CARMEN
REYNOLDSt,
MICHAEL
A.
PALLADINO,
JR.t,
AND
DAVID
V.
GOEDDEL*
Departments
of
*Molecular
Biology
and
tCell
Biology,
Genentech,
Inc.,
460
Point
San
Bruno
Boulevard,
South
San
Francisco,
CA
94080
Communicated
by
Bruce
N.
Ames,
July
25,
1991
ABSTRACT
The
individual
roles
of
the
murine
type
1
and
type
2
tumor
necrosis
factor
(TNF)
receptors
(TNF-R1
and
TNF-R2)
were
investigated
utilizing
(i)
the
strong
species
specificity
of
TNF-R2
for
murine
TNF
compared
to
human
TNF
and
(it)
agonistic
rabbit
polyclonal
antibodies
directed
against
the
individual
TNF
receptors.
Proliferation
of
mouse
thymocytes
and
the
murine
cytotoxic
T-cell
line
CT-6
is
stim-
ulated
by
murine
TNF
but
not
by
human
TNF.
Consistent
with
this
observation,
polyclonal
antibodies
directed
against
TNF-R2
induced
proliferation
in
both
of
these
cell
types,
whereas
polyclonal
antibodies
directed
against
TNF-R1
had
no
effect.
In
contrast,
cytotoxicity
in
murine
LM
cells
(which
are
sensitive
to
murine
and
human
TNF)
was
induced
by
antibodies
against
TNF-R1
but
not
by
antibodies
against
TNF-R2.
Also,
the
steady-state
level
of
manganous
superoxide
dismutase
mRNA
in
the
murine
NIH
3T3
cell
line
was
induced
by
murine
TNF,
human
TNF,
and
anti-TNF-R1
but
not
by
anti-TNF-R2.
These
results
suggest
that
TNF-R2
initiates
signals
for
the
proliferation
of
thymocytes
and
cytotoxic
T
cells,
whereas
TNF-R1
initiates
signals
for
cytotoxicity
and
the
induction
of
the
protective
activity,
manganous
superoxide
dismutase.
The
nonredundant
signaling
observed
for
the
two
TNF
receptors
cannot
be
explained
simply
by
the
differential
expression
of
the
two
TNF
receptors
in
the
various
cell
types,
because
LM
cells
express
on
their
surface
higher
levels
of
TNF-R2
than
TNF-R1,
and
LM
cells,
NIH
3T3
cells,
and
thymus
cells
all
express
mRNA
corresponding
to
both
receptor
types.
It
is
therefore
likely
that
the
two
receptors
initiate
distinct
signaling
pathways
that
result
in
the
induction
of
different
cellular
responses.
Tumor
necrosis
factor
(TNF)
is
a
multifunctional
cytokine
produced
mainly
by
activated
macrophages,
T
cells,
mast
cells,
and
some
epithelial
tumor
cells
(1-3).
The
wide
range
of
biological
effects
elicited
by
TNF
include
hemorrhagic
necrosis
of
transplanted
tumors,
growth
proliferation
of
normal
cells,
cytotoxicity,
inflammatory,
immunoregulatory
and
antiviral
responses,
and
an
important
role
in
endotoxic
shock
(4-8).
The
first
step
in
the
induction
of
these
various
cellular
responses
by
TNF
is
the
binding
to
specific
cell
surface
receptors.
TNF
receptors
have
been
detected
on
a
wide
variety
of
normal
tissues
and
cell
lines
that
are
sensitive
or
resistant
to
TNF
(9-12).
Two
immunologically
distinct
TNF
receptors
of
approximately
55
kDa
(TNF-R1)
and
75
kDa
(TNF-R2)
have
now
been
identified
(13-16),
and
human
and
mouse
cDNAs
corresponding
to
both
receptor
types
have
been
isolated
and
characterized
(17-20).
A
number
of
recent
reports
have
described
initial
studies
investigating
the
individual
roles
of
the
two
human
TNF
receptors.
Polyclonal
and
monoclonal
antibodies
directed
against
human
TNF-R1
have
been
shown
to
behave
as
receptor
agonists
and
elicit
several
TNF
activities,
such
as
cytotoxicity,
fibroblast
proliferation,
resistance
to
chlamid-
iae,
and
synthesis
of
prostaglandin
E2
(15,
21,
22).
Monoclo-
nal
antibodies
against
human
TNF-R1
that
block
the
binding
of
TNF
to
TNF-R1
and
antagonize
several
TNF
effects
have
also
been
described
(21-23).
Although
no
direct
signaling
role
for
TNF-R2
has
yet
been
identified
with
either
receptor
agonists
or
transfection
studies,
several
reports
have
de-
scribed
monoclonal
antibodies
directed
against
TNF-R2
that
can
partially
antagonize
TNF
responses
(such
as
cytotoxicity
and
activation
of
NF-KB)
and
enhance
the
antagonistic
ef-
fects
of
anti-TNF-R1
monoclonal
antibodies
(22-24).
These
reports
suggested
that
both
TNF
receptors
are
active
in
signal
transduction
and
that
there
is
redundancy
in
the
function
of
the
two
receptors.
However,
the
reported
effects
of
the
TNF-R2
antagonists
have
been
quite
small
and
were
ob-
served
exclusively
at
very
low
TNF
concentrations.
It
is
therefore
possible
that
TNF-R2
is
only
participating
as
a
minor
accessory
component
to
TNF-R1
in
the
signaling
of
these
responses.
In
this
report
we
describe
a
direct
role
of
TNF-R2
in
stimulating
proliferation
of
murine
thymocytes
and
T
cells
and
show
that
this
receptor
is
distinct
from
the
receptor
(TNF-R1)
mediating
cytotoxicity.
MATERIALS
AND
METHODS
Reagents.
Recombinant
murine
TNF
(muTNF)
and
recom-
binant
human
TNF
(hTNF)
(specific
activity
>107
units/mg)
were
provided
by
the
Genentech
manufacturing
group.
Rab-
bit
anti-murine
TNF-R1
and
rabbit
anti-murine
TNF-R2
antibodies
were
generated
against
the
soluble
extracellular
domain
of
the
corresponding
receptors
(ref.
20;
R.F.W.
and
D.V.G.,
unpublished
results).
The
titers
of
these
antisera
were
quantitated
by
a
direct
antigen-coated
ELISA.
The
dilutions
of
anti-TNF-Rl
and
anti-TNF-R2
giving
50o
bind-
ing
to
the
corresponding
purified
soluble
receptor
were
1:109,000
and
1:104,000,
respectively.
The
cross-reactive
titers
of
TNF-R1
antiserum
to
soluble
TNF-R2
and
TNF-R2
antiserum
to
soluble
TNF-R1
were
<1:200.
C3H/HeJ
mice
(The
Jackson
Laboratory)
were
used
as
the
source
of
fresh
thymocytes.
Thymocyte
Proliferation
Assay.
C3H/HeJ
thymocytes
were
cultured
in
96-well
flat-bottomed
culture
plates
(1.5
X
106
per
0.1
ml)
(Costar)
in
Eagle's
minimal
essential
medium
supplemented
with
10%t
heat-inactivated
fetal
bovine
serum
(HyClone),
1%
L-glutamine,
1%
nonessential
amino
acids,
100
units
of
penicillin
per
ml,
100
jug
of
streptomycin
per
ml,
0.1%
gentamicin
(GIBCO),
and
0.05
mM
2-mercaptoethanol
(Sigma)
in
the
presence
of
0.1%
phytohemagglutinin
P
Abbreviations:
TNF,
tumor
necrosis
factor;
muTNF,
recombinant
murine
TNF;
hTNF,
recombinant
human
TNF;
TNF-R1,
TNF
receptor
type
1;
TNF-R2,
TNF
receptor
type
2;
MnSOD,
manganous
superoxide
dismutase;
PHA-P,
phytohemagglutinin
P;
PAM-i,
plas-
minogen
activator
inhibitor
1;
IL-6,
interleukin
6.
9292
The
publication
costs
of
this
article
were
defrayed
in
part
by
page
charge
payment.
This
article
must
therefore
be
hereby
marked
"advertisement"
in
accordance
with
18
U.S.C.
§1734
solely
to
indicate
this
fact.
Proc.
Natl.
Acad.
Sci.
USA
88
(1991)
9293
(PHA-P;
Difco).
PHA-P,
muTNF,
and
antibodies
were
added
to
a
final
volume
of
0.2
ml.
After
60
h
at
370C,
cultures
were
pulsed
with
1
uCi
of
[3H]thymidine
(5
Ci/mmol;
1
Ci
=
37
GBq;
New
England
Nuclear)
for
12
h
and
harvested
onto
glass
fiber
filters
(PhD;
Cambridge
Technology,
Watertown,
MA);
mean
[3H]thymidine
incorporation
(cpm)
of
triplicate
cultures
was
determined
using
a
liquid
scintillation
counter
(Beckman).
CT-6
Proliferation
Assay.
CT-6
cells
(25)
were
cultured
in
96-well
flat-bottomed
culture
plates
(5.0
x
104
per
0.1
ml)
(Costar)
in
RPMI
medium
supplemented
with
10%o
fetal
calf
serum
(HyClone),
1%
L-glutamine,
100
units
of
penicillin
per
ml,
and
100
Ag
of
streptomycin
per
ml
(GIBCO).
muTNF
and
antisera
were
added
to
a
final
volume
of
0.2
ml.
After
24 h
at
370C,
cultures
were
pulsed
with
1
ACi
of
[3H]thymidine
(5
Ci/mmol)
for
4
h
and
harvested
onto
glass
fiber
filters,
and
mean
[3H]thymidine
incorporation
(cpm)
of
triplicate
cul-
tures
was
determined.
LM
Cytotoxicity
Assay.
LM
cells
(2
x
104
cells
per
well)
were
seeded
into
96-well
microtiter
plates
in
200
p1
of
medium
[RPMI
medium
supplemented
with
10%
fetal
calf
serum,
1%
L-glutamine,
100
units
of
penicillin
per
ml,
100
1g
of
strep-
tomycin
per
ml
(GIBCO),
and
5
1ug
of
insulin
per
ml]
and
incubated
24
h
at
370C
in
a
5%
CO2
atmosphere.
Medium
was
then
brought
to
10
pug
of
cycloheximide
per
ml
and
the
anti-TNF-R
antibodies
were
added
to
the
wells
and
serially
diluted.
The
plates
were
incubated
for
an
additional
16
h
and
the
viable
cells
were
stained
with
20%
methanol
containing
0.5%
crystal
violet.
The
dye
was
eluted
with
0.1
M
sodium
citrate
and
50%o
ethanol
and
absorbance
was
measured
at
540
nm.
Northern
Analysis.
Total
cytoplasmic
RNA
was
extracted
from
cells
as
described
(26),
electrophoresed
on
a
1.2%
form-
aldehyde/agarose
gel
(15
Ag
of
RNA
per
lane),
and
transferred
to
nitrocellulose
filters.
Filters
were
hybridized
and
washed
as
described
(26).
The
probes
used
for
filter
hybridizations
were
either
a
random-primed
32P-labeled
0.8-kilobase
(kb)
fiagment
(entire
coding
region)
of
the
manganous
superoxide
dismutase
(MnSOD)
cDNA
(27)
or
32P-labeled
oligonucleotides
corre-
sponding
to
the
murine
genes
encoding
interleukin
6
(IL-6),
plasminogen
activator
inhibitor
1
(PAI-1),
P2-microglobulin,
and
c-fos.
Autoradiography
was
done
at
-70°C
using
Kodak
intensifying
screens.
RESULTS
Thymocyte
and
T-Cell
Proliferation
Is
Stimulated
by
Anti-
bodies
to
TNF-R2.
In
a
previous
report
we
described
the
cloning
of
cDNAs
encoding
the
two
murine
TNF
receptors
and
showed
that
TNF-R2
binds
muTNF
with
high
affinity
but
fails
to
recognize
hTNF
(20).
This
observation
suggested
that
the
various
activities
of
hTNF
reported
in
mice
or
in
murine
cell
lines
would
be
mediated
by
TNF-R1,
whereas
those
activities
that
displayed
a
strong
species
specificity
for
muTNF
might
be
regulated
by
TNF-R2.
Literature
searches
revealed
that
the
majority
of
TNF
effects
reported
in
mice
or
murine
cell
lines
exhibited
little
species
preference.
An
exception
to
this
was
TNF-induced
proliferation
of
murine
thymocytes
and
the
cytotoxic
T-cell
line
CT6
(25,
28-30).
muTNF
concentrations
as
low
as
100
units/ml
(<10
ng/ml)
could
induce
a
strong
proliferative
response
in
both
of
these
cell
types,
whereas
hTNF
had
no
effect
even
at
concentra-
tions
of
>10,000
units/ml
(=1
,ug/ml).
To
examine
more
directly
the
receptor
that
mediates
proliferation
in
these
cell
types,
we
tested
highly
specific
polyclonal
antibodies
di-
rected
against
the
individual
receptors
for
possible
agonist
activities.
As
shown
in
Fig.
lA,
muTNF
stimulates
C3H/HeJ
thy-
mocyte
proliferation
in
the
presence
of
a
submitogenic
dose
of
PHA-P.
No
stimulation
is
seen
with
hTNF
even
at
doses
.2b
15-
C
E
10
o00
0C
0
-
101
1-2
103
104
muTNF
(U/ml)
12-
CCO
10-
B
°
E
6E
00C.
0C
0
-
10-6
10
10-4
10-3
102
10-1
Serum
Dilution
FIG.
1.
Proliferation
of
murine
thymocytes
in
response
to
muTNF
(A)
and
antibodies
directed
against
the
murine
TNF
recep-
tors
(B.
*,
muTNF;
*,
anti-TNF-R1;
o,
anti-TNF-R2;
e,
prebleed-
TNF-R1;
o,
prebleed-TNF-R2.
Thymocytes
were
cultured
for
60
h
with
a
submitogenic
dose
of
PHA-P
and
the
indicated
concentrations
of
muTNF
or
anti-TNF-R
antibody.
Cultures
were
then
pulse
labeled
with
[3H]thymidine
for
12
h
and
mean
[3H]thymidine
incorporation
(±SEM)
of
triplicate
cultures
was
determined.
The
amount
of
3H
incorporation
in
thymocytes
treated
with
PHA-P
alone
is
indicated
by
a dashed
line.
No
proliferation
was
observed
in
the
absence
of
PHA-P.
as
high
as
105
units/ml
(refs.
28
and
29;
data
not
shown).
Thymocyte
proliferation
was
also
examined
in
response
to
various
dilutions
of
polyclonal
antibodies
directed
against
murine
TNF-R1
and
TNF-R2.
Antibodies
to
TNF-R2
strongly
stimulated
the
proliferation
of
the
C3H1/HeJ
thymo-
cytes
even
at
a
dilution
factor
of
104.
Polyclonal
antibodies
directed
against
TNF-R1
had
no
effect
at
any
concentration
tested
despite
the
similar
titer
of
the
two
types
of
antisera
(Fig.
18).
A
similar
set
of
experiments
was
performed
with
the
interleukin
2
(IL-2)-dependent
cytotoxic
T-cell
line
CT6.
When
IL-2
is
removed
from
the
growth
medium,
muTNF
can
serve
as
a
proliferative
signal
(Fig.
2A),
whereas
hTNF
cannot
(refs.
25
and
30;
data
not
shown).
Polyclonal
anti-
bodies
against
TNF-R2
also
stimulated
the
growth
of
CT6
cells
and
the
magnitude
of
the
proliferative
response
was
similar
to
that
seen
with
muTNF
(Fig.
2B).
No
significant
effect
was
observed
with
polyclonal
antibodies
directed
against
TNF-R1.
The
agonist
activity
of
the
anti-TNF-R2
antibodies
in
the
thymocyte
and
T-cell
proliferation
assays
indicates
that
TNF-R2
can
signal
proliferation
in
at
least
some
T-cell
populations
and
also
that
TNF
is
not
required
for
the
signal
transmission.
Cytotoxicity
in
LM
Ceils
Is
Induced
by
Antibodies
to
TNF-
Rl.'One
trivial
explanation
for
the
inability
of
the
TNF-R1
antibodies
to
induce
T-cell
and
thymocyte
proliferation
was
that
these
antibodies
do
not
possess
agonist
activity
even
for
those
effects
that
are
normally
signaled
by
TNF-R1.
It
has
been
shown
previously
that
polyclonal
as
well
as
some
monoclonal
antibodies
directed
against
human
TNF-R1
can
Immunology:
Tartaglia
et
A
9294
Immunology:
Tartaglia
et
al.
)
1.
r.
10-6
10-5
104
Serum
Dilution
10-3
10-2
FIG.
2.
Proliferation
of
CT6
cells
in
response
to
muTNF
(A)
and
antibodies
directed
against
the
murine
TNF
receptors
(B).
A,
muTNF;
m,
anti-TNF-R1;
o,
anti-TNF-R2;
e,
prebleed-TNF-R1;
o,
prebleed-TNF-R2.
CT6
cells
were
cultured
for
24
h
with
the
indicated
concentrations
of
muTNF
or
anti-TNF-R
antibody.
Cultures
were
then
pulse
labeled
with
[3H]thymidine
for
4
h
and
mean
[3H]thymi-
dine
incorporation
(±SEM)
of
triplicate
cultures
was
determined.
The
amount
of
3H
incorporation
in
untreated
CT6
cells
is
indicated
by
a
dashed
line.
induce
cytotoxicity
in
human
cell
lines
(15,
21,
22),
although
no
effect
of
the
human
TNF-R1
antibodies
has
been
reported
in
murine
cell
lines.
To
determine
if
the
anti-murine
TNF-R1
antibodies
could
mimic
TNF
activity,
we
assayed
them
for
120-
100
0-
80-
60
0
Anti-muTNF-R1
>
40-
\
*
Anti-muTNF-R2
40-
20-
0.
1i-7
10-6
10-4
10-3
10-2
Serum
Dilution
FIG.
3.
Cytocidal
effect
of
anti-murine
TNF
receptor
antibodies
on
LM
cells.
o,
Anti-TNF-R1;
e,
anti-TNF-R2.
The
antisera
were
applied
for
16
h
at
the
indicated
dilutions
in
the
presence
of
cycloheximide
at
10
ug/ml.
Viability
of
cells
was
determined
(see
text).
Preimmune
sera
and
anti-NGF
antiserum
had
no
effect
in
the
range
of
serum
concentrations
used
in
this
study.
The
data
shown
are
the
mean
of
three
experiments
(±SEM).
Error
bars
have
been
omitted
for
points
with
SEM
<
the
size
of
symbol
(±2%).
cytotoxicity
against
murine
LM
cells.
LM
cells
were
chosen
because
they
possess
TNF-R1
(40%o)
and
TNF-R2
(60%o)
(20)
and
show
a
similar
sensitivity
to
muTNF
and
hTNF
(indic-
ative
of
a
TNF-R1
response)
(30).
As
shown
in
Fig.
3,
LM
cells'
were
highly
sensitive
to
antibodies
against
TNF-R1
but
not
TNF-R2.
These
results
indicate
that
the
antibodies
to
muTNF-R1
are
agonistic
and
that
TNF-R1
can
mediate
cytotoxicity
in
murine
cells.
The
resistance
of
LM
cells
to
the
TNF-R2
antibodies,
despite
the
cell
surface
expression
of
TNF-R2,
suggests
that
TNF-R2
cannot
deliver
a
cytotoxic
signal
in
murine
LM
cells.
Similar
TNF-R1
specific
cytotox-
icity
was
also
seen
with
the
murnine
cell
lines
B6MS5,
L929,
and
NIH
3T3
(data
not
shown).
TNF
Induction
of
MnSOD
mRNA
Is
Mediated
by
TNF-R1.
The
mitochondrial
enzyme
MnSOD
is
an
important
determi-
nant
of
cellular
resistance
to
the
cytotoxic
effects
of
TNF
(27).
In
addition,
MnSOD
synthesis
is
specifically
and
rapidly
induced
by
TNF
treatment
of
many
cell
types
(26).
We
therefore
tested
whether
induction
of
the
MnSOD
gene
was
mediated
by
the
same
or
different
TNF
receptor
as the
one
that
signals
cytotoxicity.
To
make
a
first
approximation
of
the
individual
roles
of
TNF-R1
and
TNF-R2
in
mediating
Mn-
SOD
induction,
we
examined
the
TNF
species
specificity
of
this
response.
As
shown
in
Fig.
4,
muTNF
and
hTNF
strongly
induced
the
1-kb
and
4-kb
MnSOD
transcripts
in
the
murine
NIH
3T3
cell
line.
Both
cytokines
also
induced
the
steady-state
mRNA
levels
of
the
genes
encoding
IL-6,
PAI-1,
.82-microglobulin,
and
c-fos.
MnSOD
mRNA
levels
were
induced
by
muTNF
and
hTNF
during
a
short
ex'osure
(3
h)
in
the
presence
of
the
protein
synthesis
inhibitor
cyclohexi-
mide
and
a
longer
exposure
(12
h)
in
the
absence
of
cyclo-
Ch
-HX
1
2h
-
C
HX
2
.
~~
MnSOD
6X
IL-6
0
w
PAl-1
2-
Microglobulin
IW
c-tos
FIG.
4.
Induction
of
mRNA
in
murine
NIH
3T3
cells.
The
left
three
lanes
show
steady-state
levels
of
mRNA
encoding
MnSOD,
IL-6,
PAI-1,
f2-microglobulin,
and
c-fos
after
a
3-h
treatment
period
in
the
presence
of
10
,g
of
cycloheximide
(CHX)
per
ml:
control
(C),
100
ng
of
muTNF
per
ml,
100
ng
of
hTNF
per
ml.
The
right
three
lanes
show
steady-state
mRNA
levels
after
a
12-h
treatment
period
in
the
absence
of
cycloheximide:
control
(C),
100
ng
of
muTNF
per
ml,
100
ng
of
hTNF
per
ml.
C
CM
.o
2
L-
x
(.)
a)
-n
E
I
°.
ovv
25
-
20
15-
10-
5-
10°
-
idi
102
103
muTNF
(U/ml)
30
CC~j
.2
6
"
x
00
0-
o
C.
'
C
C3
cis=
25
20
15
10
Proc.
Natl.
Acad
Sci.
USA
88
(1991)
Immunology:
Tartaglia
et
al.
0
z
z
0
FIG.
5.
Northern
analysis
of
MnSOD
mRNA
in
murine
NIH
3T3
cells.
Cells
were
treated
with
the
indicated
reagents
for
12
h:
control,
100
ng
of
muTNF
per
ml,
1:100
dilution
of
anti-TNF-R1
serum,
1:100
dilution
of
anti-TNF-R2
serum,
1:100
dilution
of
anti-NGF
serum.
heximide.
These
results
suggested
that
MnSOD
induction
is
signaled
through
TNF-R1.
To
test
this
prediction,
NIH
3T3
cells
were
treated
with
the
agonistic
anti-TNF-R1
and
anti-
TNF-R2
antibodies.
As
shown
in
Fig.
5,
anti-TNF-R1
anti-
bodies
strongly
induced
MnSOD
mRNA,
whereas
anti-
TNF-R2
antibodies
had
no
effect.
These
results
demonstrate
that
the
receptor
responsible
for
signaling
cytotoxicity
(TNF-
R1)
also
mediates
the
induction
of
a
key
protective
activity.
DISCUSSION
We
have
previously
shown
that
murine
TNF-R1
has
a
similar
affinity
for
muTNF
and
hTNF,
whereas
murine
TNF-R2
is
specific
for
muTNF
(20).
The
murine
system
therefore
allows
predictions
to
be
made
as
to
which
TNF
receptor
mediates
a
given
response:
TNF-R1 responses
should
be
induced
by
muTNF
and
hTNF,
whereas
TNF-R2
responses
should
only
be
induced
by
muTNF.
To
validate
this
model
and
more
directly
examine
the
individual
roles
of
the
two
TNF
recep-
tors,
we
generated
rabbit
polyclonal
antibodies
against
sol-
uble
forms
of
both
muTNF
receptors.
Polyclonal
antibodies
directed
against
TNF-R2
were
found
to
stimulate
proliferation
of
murine
thymocytes
and
the
cytotoxic
T-cell
line
CT6.
However,
polyclonal
antibodies
directed
against
TNF-R1
had
no
such
proliferative
effect.
These
results
are
consistent
with
the
proliferation
of
these
cell
types
in
response
to
muTNF,
and
not
hTNF
(25,
28-30),
which
is
also
suggestive
of
a
TNF-R2
response.
Cytotoxicity
in
murine
LM
cells
is
standardly
used
as
a
sensitive
assay
for
hTNF
(31).
Also,
LM
cells
exhibit
a
similar
sensitivity
to
muTNF
and
hTNF
(30).
This
is
sugges-
tive
of
a
response
mediated
by
TNF-R1
with
little
if
any
requirement
for
the
binding
of
TNF
to
TNF-R2.
In
agreement
with
this,
polyclonal
antibodies
directed
against
TNF-R1
induced
cytotoxicity
in
LM
cells,
even
at
a
serum
dilution
of
1:104.
No
cytotoxicity
was
seen
with
TNF-R2
antibodies,
despite
the
ability
of
these
antibodies
to
behave
as
agonists
in
the
T-cell
proliferation
assays.
It
is
interesting
to
note
that
induction
of
the
mRNA
encoding
the
defense
activity
Mn-
SOD
was
also
TNF-R1
specific.
Overexpression
of
MnSOD
mRNA
has
been
previously
shown
to
counteract
the
cyto-
toxic
effects
of
TNF,
and
therefore
mitochondrial
generation
of
has
been
implicated
as
a
key
component
of
TNF-
mediated
cell
killing
(27).
Thus,
it
appears
that
the
cascade
of
events
that
lead
to
generation
of
01
and
the
induction
of
a
02
scavaging
activity
are
signaled
by
the
same
TNF
receptor.
The
inability
of
the
TNF-R2
antibodies
to
act
as
agonists
in
the
LM
cytotoxicity
assay
was
not
due
to
the
absence
of
TNF-R2
on
LM
cells;
we
previously
showed
that
the
TNF
Proc.
Natl.
Acad.
Sci.
USA
88
(1991)
9295
receptors
expressed
on
the
surface
of
LM
cells
are
about
60%
TNF-R2
and
40%
TNF-R1
(20).
In
addition,
the
NIH
3T3
cells
used
in
the
MnSOD
induction
experiments
expressed
transcripts
corresponding
to
TNF-R1
and
TNF-R2
(data
not
shown).
The
results
of
the
cytotoxicity
and
MnSOD
mRNA
studies
therefore
suggest
that
the
functions
of
TNF-R1
and
TNF-R2
are
not
redundant,
but
rather
that
only
TNF-R1
signals
these
two
responses.
A
similar
argument
can
be
made
for
the
different
behavior
of
the
two
antibody
preparations
in
the
T-cell
and
thymocyte
proliferation
assays.
Whereas
CT6
cells
express
little
or
no
TNF-R1
mRNA
and
do
not
bind
detectable
amounts
of
hTNF
(20),
thymus
cells
express
mRNA
for
both
receptor
types
(20)
and
the
thymocytes
used
in
the
proliferation
assays
bind
muTNF
and
hTNF
(29)
(indicative
of
the
presence
of
TNF-
Ri).
Therefore,
it
would
appear
that
the
proliferative
signal
for
these
cells
can
be
mediated
by
TNF-R2
but
not
by
TNF-R1.
These
results
indicate
that
different
TNF
receptors
signal
cytotoxicity
and
T-cell
proliferation
and
that
the
two
TNF
receptors
are
not
redundant
in
signaling
these
functions.
The
amount
of
primary
sequence
similarity
between
TNF-R1
and
TNF-R2
is
also
suggestive
of
distinct
functions
for
the
two
TNF
receptors.
For
although
the
extracellular
ligand-binding
domains
of
the
two
TNF
receptors
show
some
homology
(=20%),
their
intracellular
domains
show
no
sig-
nificant
similarities
(20).
This
would
be
consistent
with
the
intracellular
regions
of
the
two
receptors
being
coupled
to
different
signal
transduction
pathways.
Several
reports
have
described
antibodies
directed
against
human
TNF-R1
that
have
agonist
properties
(15,
21,
22).
These
studies
demonstrate
that
a
specific
conformational
change
induced
by
the
TNF
molecule
itself
is
probably
not
responsible
for
the
activation
of
TNF-R1.
Rather,
a
nonspe-
cific
perturbation,
most
likely
receptor
dimerization
or
ag-
gregation,
can
be
sufficient
to
signal
through
this
receptor.
In
this
report,
we
show
that
TNF-R2
can
also
be
activated
by
immunoglobulins
in
the
absence
of
TNF.
Therefore,
signaling
via
TNF-R2
does
not
have
an
absolute
requirement
for
TNF
and
may
be
initiated
through
a
mechanism
similar
to
that
utilized
by
TNF-R1.
The
absence
of
reports
on
TNF-R2
agonists
may
be
a
result
of
direct
TNF-R2
responses
being
much
less
numerous
than
TNF-R1
responses.
In
support
of
this,
most
activity
com-
parisons
of
hTNF
and
muTNF
in
mice
or
murine
cell
lines
show
relatively
small
differences,
with
T-cell
and
thymocyte
proliferation
being
the
most
dramatic
exceptions.
Why
TNF-R2
effects
would
be
specific
for
such
a
small
cell
population
is
not
clear,
given
the
near
ubiquitous
distribution
of
this
receptor
in
cell
types
and
tissues.
Perhaps
a
number
of
TNF-R2
responses
are
yet
to
be
identified.
Also,
TNF-R2
may
play
an
accessory
role
in
mediating
other
TNF
effects,
even
if
it
is
not
responsible
for
the
signal
transmission.
The
experiments
described
in
this
study
show
that
TNF-R1
can
initiate
signals
for
cytotoxicity
and
TNF-R2
can
initiate
signals
for
thymocyte
and
cytotoxic
T-cell
proliferation.
However,
it
should
be
noted
that
TNF-induced
proliferation
may
not
be
mediated
by
TNF-R2
in
all
cell
types.
Engelmann
et
al.
(15)
have
shown
that
polyclonal
antibodies
against
human
TNF-R1
can
stimulate
proliferation
of
human
FS11
fibroblasts.
Thus,
it
appears
that
different
TNF
receptors
can
signal
proliferation
in
different
cell
types.
It
is
therefore
likely
that
many
additional
studies
will
be
required
before
a
thor-
ough
understanding
of
the
individual
roles
of
the
two
TNF
receptors
will
be
realized.
We
thank
Bill
Kohr
and
Helga
Raab
for
help
and
advice
on
the
purification
of
the
soluble
TNF
receptors
and
also
Greg
Bennett
and
Roderick
Vitangcol
for
preparation
of
the
polyclonal
antisera.
92%
Immunology:
Tartaglia
et
al.
1.
Decker,
T.,
Lohmann-Matthes,
M.
L.
&
Gifford,
G.
E.
(1987)
J.
Immunol.
138,
957-962.
2.
Turner,
M.,
Londei,
M.
&
Feldmann,
M.
(1987)
Eur.
J.
Immunol.
17,
1807-1814.
3.
Spriggs,
D.
R.,
Imamura,
K.,
Rodriguez,
C.,
Sariban,
E.
&
Kufe,
D.
W.
(1988)
J.
Clin.
Invest.
81,
455-460.
4.
Carswell,
E.
A.,
Old,
L.
J.,
Kassel,
R.
L.,
Green,
S.,
Fiore,
N.
&
Williamson,
B.
(1975)
Proc.
Natl.
Acad.
Sci.
USA
72,
3666-3670.
5.
Sugarman,
B.
J.,
Aggarwal,
B.
B.,
Hass,
P.
E.,
Figari,
I.
S.,
Palladino,
M.
A.
&
Shepard,
H.
M.
(1985)
Science
230,
943-
945.
6.
Goeddel,
D.
V.,
Aggarwal,
B.
B.,
Gray,
P.
W.,
Leung,
D.
W.,
Nedwin,
G.
E.,
Palladino,
M.
A.,
Patton,
J.
S.,
Pennica,
D.,
Shepard,
H.
M..
Sugarman,
B.
J.
&
Wong,
G.
H.
W.
(1986)
Cold
Spring
Harbor
Symp.
Quant.
Biol.
5,
597-609.
7.
Beutler,
B.
&
Cerami,
A.
(1988)
Annu.
Rev.
Biochem.
57,
505-518.
8.
Old,
L.
J.
(1988)
Sci.
Am.
258(5),
59-75.
9.
Kull,
F.
C.,
Jacobs,
S.
&
Cuatrecasas,
P.
(1985)
Proc.
Natl.
Acad.
Sci.
USA
82,
5756-5760.
10.
Baglioni,
C.,
McCandless,
S.,
Tavernier,
J.
&
Fiers,
W.
(1985)
J.
Biol.
Chem.
266,
13395-13397.
11.
Creasey,
A.
A.,
Yamamoto,
R.
&
Vitt,
C.
R.
(1987)
Proc.
Nat!.
Acad.
Sci.
USA
84,
3293-3297.
12.
Tsujimoto,
M.,
Yip,
Y.
K.
&
Vilcek,
J.
(1985)
Proc.
Natl.
Acad.
Sci.
USA
82,
7626-7630.
13.
Hohmann,
H.
P.,
Remy,
R.,
Brockhaus,
M.
&
van
Loon,
A.
P.
G.
M.
(1989)
J.
Biol.
Chem.
264,
14927-14934.
14.
Engelmann,
H.,
Novick,
D.
&
Wallach,
D.
(1990)
J.
Biol.
Chem.
265,
1531-1536.
15.
Engelmann,
H.,
Holtmann,
H.,
Brakebush,
C.,
Avni,
Y.
S.,
Sarov,
I.,
Nophar,
Y.,
Hadas,
E.,
Leitner,
0.
&
Wallach,
D.
(1990)
J.
Biol.
Chem.
265,
14497-14504.
16.
Brockhaus,
M.,
Schoenfeld,
H.
J.,
Schlaeger,
E.
J.,
Hun-
zicker,
W.,
Lesslauer,
W.
&
Loetscher,
H.
(1990)
Proc.
Natl.
Acad.
Sci.
USA
87,
3127-3131.
17.
Schall,
T.
J.,
Lewis,
M.,
Koller,
K.
J.,
Lee,
A.,
Rice,
G.
C.,
Wong,
G.
H.
W.,
Gatanaga,
T.,
Granger,
G.
A.,
Lentz,
R.,
Raab,
H.,
Kohr,
W.
J.
&Goeddel,
D.
V.
(1990)
Cell61,
361-370.
18.
Loetscher,
H.,
Pan,
Y.
C.
E.,
Lahm,
H.
W.,
Gentz,
R.,
Brock-
haus,
M.,
Tabuchi,
H.
&
Lesslauer,
W.
(1990)
Cell
61,
351-359.
19.
Smith,
C.
A.,
Davis,
T.,
Anderson,
D.,
Solam,
L.,
Beckmann,
M.
P.,
Jerzy,
R.,
Dower,
S.
K.,
Cosman,
D.
&
Goodwin,
R.
G.
(1990)
Science
248,
1019-1023.
20.
Lewis,
M.,
Tartaglia,
L.
A.,
Lee,
A.,
Bennett,
G.
L.,
Rice,
G.
C.,
Wong,
G.
H.
W.,
Chen,
E.
Y.
&
Goeddel,
D.
V.
(1991)
Proc.
Nat!.
Acad.
Sci.
USA
88,
2830-2834.
21.
Espevik,
T.,
Brockhaus,
M.,
Loetscher,
H.,
Nonstad,
U.
&
Shalaby,
R.
(1990)
J.
Exp.
Med.
171,
415-426.
22.
Shalaby,
M.
R.,
Sundan,
A.,
Loetscher,
H.,
Brockhaus,
M.,
Lesslauer,
W.
&
Espevik,
T.
(1990)
J.
Exp.
Med.
172,
1517-
1520.
23.
Naume,
B.,
Shalaby,
R.,
Lesslauer,
W.
&
Espevik,
T.
(1991)
J.
Immunol.
146,
3045-3048.
24.
Hohmann,
H.
P.,
Brockhaus,
M.,
Baeuerle,
P.
A.,
Remy,
R.,
Kolbeck,
R.
&
van
Loon,
A.
P.
G.
M.
(1990)
J.
Biol.
Chem.
265,
22409-22417.
25.
Ranges,
G.
E.,
Bombara,
M.
P.,
Aiyer,
R.
A.,
Rice,
G.
C.
&
Palladino,
M.
A.
(1989)
J.
Immunol.
142,
1203-1208.
26.
Wong,
G.
H.
W.
&
Goeddel,
D.
V.
(1988)
Science
242,
941-
944.
27.
Wong,
G.
H.
W.,
Elwell,
J.
H.,
Oberley,
L.
W.
&
Goeddel,
D.
V.
(1989)
Cell
58,
923-931.
28.
Ehrke,
M.
J.,
Ho,
R.
L.
X.
&
Hori,
K.
(1988)
Cancer
Immunol.
Immunother.
27,
103-108.
29.
Ranges,
G.
E.,
Zlotnik,
A.,
Espevik,
T.,
Dinarello,
C.
A.,
Cerami,
A.
&
Palladino,
M.
A.
(1988)
J.
Exp.
Med.
167,
1472-1478.
30.
Rice,
G.
C.,
Kramer,
S.
M.,
Figari,
1.
S.,
Ranges,
G.
E.
&
Palladino,
M.
A.
(1990)
in
Tumor
Necrosis
Factor:
Structure,
Mechanism
of
Action,
Role
in
Disease
and
Therapy,
eds.
Bonavida
B.
&
Granger,
G.
(Karger,
Basel),
pp.
87-93.
31.
Kramer,
S.
M.
&
Carver,
M.
E.
(1986)
J.
Immunol.
Methods
93,
201-206.
Proc.
Natl.
Acad.
Sci.
USA
88
(1991)
... In support of this, a lack of functional TNF in mice has been shown to attenuate CaCl 2− induced aneurysm development (16). TNF exists in two bioactive forms (17), but it is unclear whether both forms contribute to AAA development. TNF is synthesized as a transmembrane protein (tmTNF) (18) that is cleaved from the cell membrane by metalloproteinase TNF-converting enzyme (TACE/ADAM17) and released to the circulation and surrounding tissue as a soluble trimer complex (solTNF) (19). ...
... TNF is synthesized as a transmembrane protein (tmTNF) (18) that is cleaved from the cell membrane by metalloproteinase TNF-converting enzyme (TACE/ADAM17) and released to the circulation and surrounding tissue as a soluble trimer complex (solTNF) (19). The effect of TNF is mediated by TNF receptor (TNFR) 1 and TNFR2, resulting in both overlapping and distinct biological outcomes (17). SolTNF has a higher affinity for TNFR1, and thus primarily drives a proinflammatory response through TNFR1 activation (20), whereas tmTNF acts in a paracrine fashion and mainly signal through TNFR2, resulting in tissue repair and regeneration as well as immune-regulating functions (21)(22)(23)(24). ...
Article
Full-text available
Background Tumor necrosis factor (TNF) is pathologically elevated in human abdominal aortic aneurysms (AAA). Non-selective TNF inhibition-based therapeutics are approved for human use but have been linked to several side effects. Compounds that target the proinflammatory soluble form of TNF (solTNF) but preserve the immunomodulatory capabilities of the transmembrane form of TNF (tmTNF) may prevent these side effects. We hypothesize that inhibition of solTNF signaling prevents AAA expansion.Methods The effect of the selective solTNF inhibitor, XPro1595, and the non-selective TNF inhibitor, Etanercept (ETN) was examined in porcine pancreatic elastase (PPE) induced AAA mice, and findings with XPro1595 was confirmed in angiotensin II (ANGII) induced AAA in hyperlipidemic apolipoprotein E (Apoe) –/– mice.ResultsXPro1595 treatment significantly reduced AAA expansion in both models, and a similar trend (p = 0.06) was observed in PPE-induced AAA in ETN-treated mice. In the PPE aneurysm wall, XPro1595 improved elastin integrity scores. In aneurysms, mean TNFR1 levels reduced non-significantly (p = 0.07) by 50% after TNF inhibition, but the histological location in murine AAAs was unaffected and similar to that in human AAAs. Semi-quantification of infiltrating leucocytes, macrophages, T-cells, and neutrophils in the aneurysm wall were unaffected by TNF inhibition. XPro1595 increased systemic TNF levels, while ETN increased systemic IL-10 levels. In ANGII-induced AAA mice, XPro1595 increased systemic TNF and IL-5 levels. In early AAA development, proteomic analyses revealed that XPro1595 significantly upregulated ontology terms including “platelet aggregation” and “coagulation” related to the fibrinogen complex, from which several proteins were among the top regulated proteins. Downregulated ontology terms were associated with metabolic processes.Conclusion In conclusion, selective inhibition of solTNF signaling reduced aneurysm expansion in mice, supporting its potential as an attractive treatment option for AAA patients.
... TNFR1 is expressed by most cell types and has a higher affinity for solTNF than for tmTNF, leading to classical nuclear factor-kappa B (NF-κB)-mediated transcription of inflammatory genes, but it also contains a death domain that can either lead to caspase-mediated apoptosis or caspase-independent necroptosis [14][15][16]. TNFR2 is preferentially found on immune cells, glial cells, and endothelial cells, is primarily activated upon binding of tmTNF, and is linked to immune modulation and cell proliferation and survival [14,17]. ...
Article
Full-text available
Clinical and animal model studies have implicated inflammation and glial and peripheral im-mune cell responses in the pathophysiology of spinal cord injury (SCI). A key player in the in-flammatory response after SCI is the pleiotropic cytokine tumor necrosis factor (TNF), which ex-ists both in both transmembrane (tmTNF) and a soluble (solTNF) form. In the present study, we extend our previous findings of a therapeutic effect of topically blocking solTNF signaling after SCI for three consecutive days on lesion size and functional outcome to study the effect on spatio-temporal changes in the inflammatory response after SCI in mice treated with the selective solTNF inhibitor XPro1595 and compared to saline-treated mice. We found that despite compa-rable TNF and TNF receptor levels between XPro1595- and saline-treated mice, XPro1595 transi-ently decreased pro-inflammatory interleukin (IL)-1 and IL-6 levels and increased pro-regenerative IL-10 levels in the acute phase after SCI. This was complemented by a decrease in the number of infiltrated leukocytes (macrophages and neutrophils) in the lesioned area of the spinal cord and an increase in the number of microglia in the peri-lesion area 14 days after SCI, fol-lowed by a decrease in microglial activation in the peri-lesion area 21 days after SCI. This trans-lated into increased myelin preservation and improved functional outcomes in XPro1595-treated mice 35 days after SCI. Collectively, our data suggests that selective targeting of solTNF time-dependently modulates the neuroinflammatory response by favoring a pro-regenerative envi-ronment in the lesioned spinal cord, leading to improved functional outcomes.
... mTNF-TNFR2 binding generates a more effective response than sTNF [38]. TNFR1 and TNFR2 show different intracellular structures that bind several adaptor proteins [39]. The TNFR1 cytoplasmic tail includes the death domain (DD), thus leading it to engage the TNFR1-associated DD (TRADD) [40]; by comparison, TNFR2 recruits TNFR-associated factor (TRAF) 1 and 2 proteins [41]. ...
Article
Full-text available
Ischemic heart disease is the principal cause of death worldwide and clinically manifests as myocardial infarction (MI), stable angina, and ischemic cardiomyopathy. Myocardial infarction is defined as an irreversible injury due to severe and prolonged myocardial ischemia inducing myocardial cell death. Revascularization is helpful in reducing loss of contractile myocardium and improving clinical outcome. Reperfusion rescues myocardium from cell death but also induces an additional injury called ischemia-reperfusion injury. Multiple mechanisms are involved in ischemia-reperfusion injury, such as oxidative stress, intracellular calcium overload, apoptosis, necroptosis, pyroptosis, and inflammation. Various members of the tumor necrosis factor family play a key role in myocardial ischemia-reperfusion injury. In this article, the role of TNFα, CD95L/CD95, TRAIL, and the RANK/RANKL/OPG axis in the regulation of myocardial tissue damage is reviewed together with their potential use as a therapeutic target.
... In fact, it became clear that two different receptors existed that bound TNF as well as LTα yet displayed functional differences [34][35][36]. Only one receptor termed TNF-Receptor-1 (TNFR-1) (p55) was shown to be important for the protection against bacterial infection and the proinflammatory activity of endotoxins [37][38][39], while the role of the second TNFR-2 p75 remains to some extent unresolved to this day, despite a demonstrated function in cell proliferation [40], regulatory T cell proliferation [2], and general support for p55 [41,42]. The immediate TNF family (TNF and LTa) was extended with the discovery of a new partner of LTα, LTβ [43,44]. ...
Article
Full-text available
Tumor Necrosis Factor was one of the first cytokines described in the literature as a soluble mediator of cytotoxicity to tumors. Over the years, more extensive research that tried to employ Tumor Necrosis Factor in cancer treatments showed nevertheless that it mainly functioned as a proinflammatory cytokine. However, this did not stop the search for the holy grail of cancer research: A cytokine that could act as a one-stop treatment for solid tumors and lymphomas. This review will summarize the long experimental history of Tumor Necrosis Factor that caused the initial observations of a tumor necrotizing cytokine that could serve as a potential cancer treatment and discuss the current state of research into this side of the activities of Tumor Necrosis Factor.
... During M.tb infection, TNF-α is one of the earliest cytokines to be produced. TNF-α signals through two trimeric membrane receptors: TNF receptors 1(TNF-R1) and (TNF-R2) [140] or by reverse signaling back into the membrane of TNFproducing cells [141]. Consequently, TNF signaling can result in a diversity of biological functions. ...
Article
Full-text available
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Article
Full-text available
Cellular responses to TNF are inherently heterogeneous within an isogenic cell population and across different cell types. TNF promotes cell survival by activating pro-inflammatory NF-κB and MAPK signalling pathways but may also trigger apoptosis and necroptosis. Following TNF stimulation, the fate of individual cells is governed by the balance of pro-survival and pro-apoptotic signalling pathways. To elucidate the molecular mechanisms driving heterogenous responses to TNF, quantifying TNF/TNFR1 signalling at the single-cell level is crucial. Fluorescence live-cell imaging techniques offer real-time, dynamic insights into molecular processes in single cells, allowing for detection of rapid and transient changes, as well as identification of subpopulations, that are likely to be missed with traditional endpoint assays. Whilst fluorescence live-cell imaging has been employed extensively to investigate TNF-induced inflammation and TNF-induced cell death, it has been underutilised in studying the role of TNF/TNFR1 signalling pathway crosstalk in guiding cell-fate decisions in single cells. Here, we outline the various opportunities for pathway crosstalk during TNF/TNFR1 signalling and how these interactions may govern heterogenous responses to TNF. We also advocate for the use of live-cell imaging techniques to elucidate the molecular processes driving cell-to-cell variability in single cells. Understanding and overcoming cellular heterogeneity in response to TNF and modulators of the TNF/TNFR1 signalling pathway could lead to the development of targeted therapies for various diseases associated with aberrant TNF/TNFR1 signalling, such as rheumatoid arthritis, metabolic syndrome, and cancer.
Article
Central tolerance depends upon Ag-mediated cell death in developing thymocytes. However, the mechanism of induced death is poorly understood. Among the known death-inducing proteins, TNF was previously found to be constitutively expressed in the thymus. The role of TNF in thymocyte negative selection was therefore investigated using TNF receptor (TNFR)-deficient mice containing a TCR transgene. TNFR-deficient mice displayed aberrant negative selection in two models: an in vitro system in which APC are cultured with thymocytes, and a popular in vivo system in which mice are treated with anti-CD3 Abs. In contrast, TNFR-deficient mice displayed normal thymocyte deletion in two Ag-induced in vivo models of negative selection. Current models of negative selection and the role of TNFR family members in this process are discussed in light of these results.
Article
An arsenal of molecular tools with increasingly diversified mechanisms of action is being developed by the scientific community to enable biological interrogation and pharmaceutical modulation of targets and pathways of ever increasing complexity. While most small molecules interact with the target of interest in a 1 : 1 relationship, a noteworthy number of recent examples were reported to bind in a sub-stoichiometric manner to a homomeric protein complex. This approach requires molecular understanding of the physiologically relevant protein assemblies and in-depth characterization of the compound's mechanism of action. The recent literature examples summarized here were selected to illustrate methods used to identify and characterize molecules with such mechanisms. The concept of one small molecule targeting a homomeric protein assembly is not new but the subject deserves renewed inspection in light of emerging technologies and increasingly diverse target biology, to ensure relevant in vitro systems are used and valuable compounds with potentially novel sub-stoichiometric mechanisms of action aren't overlooked.
Article
Full-text available
: Tumor necrosis factor-alpha (TNF-α) is a pro-inflammatory cytokine implicated in various physiological and pathological events. Carnitine is a quaternary amine which plays a significant role in fatty acid oxidation and is reported to produce antiapoptotic effects. Aim of this work was to study the effect of L-Carnitine (LC) on TNF-α induced apoptosis in mice oocytes. : Oocytes were isolated from super ovulated Swiss Albino mice and treated with different concentrations of TNF-α (0.1ng/ml, 1ng/ml, 10ng/ml, 100ng/ml) and LC (0.1mg/ml, 0.3mg/ml, 0.5mg/ml, 1.0 mg/ml). TUNEL Assay was done for the biochemical assessment of apoptosis. : Apoptotic indices with different doses of TNF-α (0.1ng/ml, 1ng/ml, 10ng/ml, 100 ng/ml) were 28.5%, 71.4%, 100%, 42.8% respectively. The concentration of TNF-α that produced the highest apoptotic index was 10ng/ml. LC alone in different doses (0.1mg/ml, 0.3mg/ml, 0.5mg/ml, 1.0 mg/ml) did not elicit any apoptotic signal. Further LC was added in different doses with 10ng/ml TNF-α to study the rate of apoptosis in mice oocytes. Apoptotic index with 10 ng/ml TNF-α and different doses of LC (0.1mg/ml, 0.3mg/ml, 0.5mg/ml, 1 mg/ml) were 25%, 37.5%, 50%, 62.5% respectively. The concentration of LC that reduced the apoptotic index to the maximum was 0.1mg/ml. : Present study could demonstrate the anti-apoptotic effect of LC against apoptotic effects of TNF-α in mice oocytes. The study presents preliminary data suggesting a possible therapeutic role of LC in inflammatory etiologies such as ovarian failure.
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
Immunological cross-reactivity between tumor necrosis factor (TNF) binding proteins which are present in human urine (designated TBPI and TBPII) and two molecular species of the cell surface receptors for TNF is demonstrated. The two TNF receptors are shown to be immunologically distinct, to differ in molecular weight (58,000 and 73,000), and to be expressed differentially in different cells. It is further shown that polyclonal antibodies against one of the TNF binding proteins (TBPI) display, by virtue of their ability to bind the TNF receptor, activities which are very similar to those of TNF. These antibodies are cytotoxic to cells which are sensitive to TNF toxicity, induce resistance to TNF toxicity, enhance the incorporation of thymidine into normal fibroblasts, inhibit the growth of chlamydiae, and induce the synthesis of prostaglandin E2. Monovalent F(ab) fragments of the polyclonal antibodies lack TNF-like activities, but acquire them upon cross-linking with anti-F(ab)2 antibodies, suggesting that the ability of the anti-TBPI antibodies to mimic TNF correlates with their ability to cross-link the TNF receptors. This notion was further supported by data obtained in a comparative study of the TNF-like cytotoxicity of a panel of monoclonal antibodies against TBPI. The induction of TNF-like effects by antibodies to a TNF receptor suggests that TNF is not directly involved in intracellular signalling. Rather, it is the receptors to this cytokine which, when properly triggered in a process which appears to involve clustering of these receptors, transduce the signal for response to TNF into the cell's interior.
Article
Full-text available
Complementary DNA clones encoding two distinct tumor necrosis factor receptors were isolated from a mouse macrophage cDNA library. The cDNA for murine tumor necrosis factor receptor type 1 (mTNF-R1) predicts a mature polypeptide of 425 amino acids that is 64% identical to its human counterpart, whereas the cDNA of murine tumor necrosis factor receptor type 2 (mTNF-R2) predicts a mature protein of 452 amino acids that is 62% identical to human tumor necrosis factor receptor type 2. The two murine tumor necrosis factor receptors have limited sequence homology (approximately 20% identity) in their extracellular regions but no apparent similarity in their cytoplasmic portions. Northern (RNA) analysis indicates a single 2.6-kilobase (kb) transcript for mTNF-R1; a 3.6-kb and a more predominant 4.5-kb transcript are observed for mTNF-R2. A human cell line transfected with either mTNF-R1 or mTNF-R2 expression vectors specifically bound 125I-labeled recombinant murine tumor necrosis factor alpha (TNF-alpha). Although mTNF-R1 had a similar affinity for both recombinant murine TNF-alpha and human TNF-alpha, mTNF-R2 showed strong specificity for recombinant murine TNF-alpha. This result suggests that the various activities of human tumor necrosis factor alpha reported in mice or in murine cell lines are probably mediated by mTNF-R1.
Article
Full-text available
Two proteins which specifically bind tumor necrosis factor (TNF) were isolated from human urine by ligand (TNF)-affinity purification, followed by reversed phase high performance liquid chromatography. The molecular weights of the two proteins, as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, were similar (about 30,000). Both proteins provided protection against the cytocidal effect of TNF in vitro and both bound TNF-alpha more effectively than TNF-beta. Antibodies raised against each of the proteins had an inhibitory effect on the binding of TNF to cells, suggesting that both proteins are structurally related to the TNF receptors. However, the two proteins differed in NH2-terminal amino acid sequences: Asp-Ser-Val-Cys-Pro- in one and Val-Ala-Phe-Thr-Pro- in the other. The NH2-terminal sequence of the former protein was invariable, while that of the latter was truncated to varying degrees. The two proteins were also immunologically distinct. The relative efficacy of anti-sera against the two proteins in inhibiting the binding of TNF to cells varied markedly from one line of cells to another. Evidence has been presented recently for the existence of two distinct molecular species of cell surface receptors for TNF and for differential expression of those two receptors by cells of different lines. The findings presented in this study are consistent with the notion that the urinary TNF-binding proteins constitute soluble forms of the two molecular species of the cell surface TNF receptors.
Article
In studying "hemorrhagic necrosis" of tumors produced by endotoxin, it was found that the serum of bacillus Calmette--Guerin (BCG)-infected mice treated with endotoxin contains a substance (tumor necrosis factor; TNF) which mimics the tumor necrotic action of endotoxin itself. TNF-positive serum is as effective as endotoxin itself in causing necrosis of the sarcoma Meth A and other transplanted tumors. A variety of tests indicate that TNF is not residual endotoxin, but a factor released from host cells, probably macrophages, by endotoxin. Corynebacteria and Zymosan, which like BCG induce hyperplasia of the reticulo-endothelial system, can substitute for BCG in priming mice for release of TNF by endotoxin. TNF is toxic in vitro for two neoplastic cell lines; it is not toxic for mouse embryo cultures. We propose that TNF mediates endotoxin-induced tumor necrosis, and that it may be responsible for the suppression of transformed cells by activated macrophages.
Article
In this study we investigated the expression of the 55 kDa (p55) and the 75 kDa (p75) TNF receptors in CD56+ NK cells and their role in NK and lymphokine-activated killer cells cell functions. By using mAb against the p55 and p75 TNF-R, NK cells were found to express both p55 and p75 upon activation, and both receptors were involved in the generation of lymphokine-activated killer cells activity. Proliferative activity of IL-2 stimulated NK cells was inhibited by anti-TNF-alpha mAb, indicating that endogenously produced TNF-alpha is important for optimal proliferation of NK cells. Furthermore, addition of rTNF-alpha increased the IL-2-induced proliferation of NK cells. mAb to p55 and p75 inhibited the IL-2-induced proliferation indicating that both TNF-R are involved in mediating this effect.
Article
The pleiotropic cyto/lymphokine tumor necrosis factor (TNF) exerts its functions by binding to specific cell-surface receptors. We have prepared two sets of monoclonal antibodies (mAbs) against TNF-binding proteins from the HL-60 (htr-mAb series) and U-937 (utr-mAb series) cell lines. The htr antibodies inhibit the binding of 125I-labeled TNF-alpha to HL-60 cells only partially, whereas they block the TNF-alpha binding to several adenocarcinoma cell lines (HEp-2, HeLa, and MCF7) almost completely. In contrast, the utr antibodies have no effect on TNF-alpha binding to the adenocarcinoma cell lines but partially inhibit TNF-alpha binding to HL-60 and U-937 cells. However, htr-9 and utr-1 antibodies in combination fully inhibit the TNF-alpha binding to HL-60 and U-937 cells. The binding of TNF-beta to HEp-2 and U-937 cells is also inhibited by htr and utr antibodies. Neither htr nor utr mAb has an effect on the TNF-sensitive murine cell lines L929 and WEHI 164. Flow cytometry studies show that mAbs htr-9 and utr-1 detect two distinct TNF-binding sites on human cell lines. Immunologic blot and immunoprecipitation analyses indicate that mAbs htr-9 and utr-1 recognize proteins of approximately 55 kDa and 75 kDa, respectively. These data provide evidence for the existence of two distinct TNF receptor molecules that contribute to varying extent to the TNF binding by different human cells.
Article
A human tumor necrosis factor (TNF) binding protein from serum of cancer patients was purified to homogeneity and partially sequenced. Synthetic DNA probes based on amino acid sequence information were used to isolate cDNA clones encoding a receptor for TNF. The TNF receptor (TNF-R) is a 415 amino acid polypeptide with a single membrane-spanning region. The extracellular cysteine-rich domain of the TNF-R is homologous to the nerve growth factor receptor and the B cell activation protein Bp50. Human embryonic kidney cells transfected with a TNF-R expression vector specifically bind both 125I-labeled and biotinylated TNF-alpha. Unlabeled TNF-alpha and TNF-beta were equally effective at displacing the binding of labeled TNF-alpha to TNF-R expressing cells. Northern analysis indicates a single species of mRNA for the TNF-R in a variety of cell types. Therefore, the soluble TNF binding protein found in human serum is probably proteolytically derived from the TNF-R.
Article
Two distinct receptors for tumor necrosis factor (TNF) of 55 and 75 kd are expressed at low levels by various cells. The 55 kd TNF receptor was purified from HL60 cells, and partial amino acid sequences were determined. Short degenerate sense and antisense oligonucleotide primers encoding the N- and C-terminal ends of a peptide of 22 amino acid residues were used to amplify a 66 bp cDNA fragment from HL60 RNA by reverse transcriptase-polymerase chain reaction. The cDNA fragment as a probe identified several overlapping clones in a human placenta cDNA library. The open reading frame of the cDNA predicts a 455 amino acid TNF receptor protein with leader, extracellular, transmembrane, and intracellular domains. When expressed in COS-1 cells or in a baculovirus system, the cDNA conferred TNF binding properties comparable to the native receptor. Surprisingly, the 55 kd TNF receptor shows a high degree of sequence homology to the NGF receptor extracellular domain.
Article
Tumor necrosis factor alpha and beta (TNF-alpha and TNF-beta) bind surface receptors on a variety of cell types to mediate a wide range of immunological responses, inflammatory reactions, and anti-tumor effects. A cDNA clone encoding an integral membrane protein of 461 amino acids was isolated from a human lung fibroblast library by direct expression screening with radiolabeled TNF-alpha. The encoded receptor was also able to bind TNF-beta. The predicted cysteine-rich extracellular domain has extensive sequence similarity with five proteins, including nerve growth factor receptor and a transcriptionally active open reading frame from Shope fibroma virus, and thus defines a family of receptors.