ArticlePDF Available

Proteasome Inhibition Results in TRAIL Sensitization of Primary Keratinocytes by Removing the Resistance-Mediating Block of Effector Caspase Maturation

Authors:

Abstract and Figures

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exerts potent cytotoxic activity against transformed keratinocytes, whereas primary keratinocytes are relatively resistant. In several cell types, inhibition of the proteasome sensitizes for TRAIL-induced apoptosis by interference with NF-kappaB activation. Here we describe a novel intracellular mechanism of TRAIL resistance in primary cells and how this resistance is removed by proteasome inhibitors independent of NF-kappaB in primary human keratinocytes. This sensitization was not mediated at the receptor-proximal level of TRAIL DISC formation or caspase 8 activation but further downstream. Activation of caspase 3 was critical, as it only occurred when mitochondrial apoptotic pathways were activated, as reflected by Smac/DIABLO, HtrA2, and cytochrome c release. Smac/DIABLO and HtrA2 are needed to release the X-linked inhibitor-of-apoptosis protein (XIAP)-mediated block of full caspase 3 maturation. XIAP can effectively block caspase 3 maturation and, intriguingly, is highly expressed in primary but not in transformed keratinocytes. Ectopic XIAP expression in transformed keratinocytes resulted in increased resistance to TRAIL. Our data suggest that breaking of this resistance via proteasome inhibitors, which are potential anticancer drugs, may sensitize certain primary cells to TRAIL-induced apoptosis and could thereby complicate the clinical applicability of a combination of TRAIL receptor agonists with proteasome inhibitors.
TRAIL dose-dependently enhances NF- ␬ B binding in primary keratinocytes. (A) Primary keratinocytes were treated with 5 to 1,000 ng of TRAIL per ml for 6 h and analyzed for NF- ␬ B DNA binding by electrophoretic mobility shift assay with nuclear extracts. Induction of p65/p50 heterodimers (upper arrow) or p50 homodimers (lower arrow) is shown. As a control for the speci fi city of DNA binding, control experiments with an excess of unlabeled probe were performed (not shown). Cell viability was determined in parallel experiments by crystal violet staining and is indicated as a percentage of the control value. (B) Primary keratinocytes were exposed to 250 ng of TRAIL per ml for 6 h in the presence or absence of 40 ␮ M zVAD-FMK. Survival of cells was determined in parallel experiments as described for A. (C) Composition of TRAIL-induced nuclear NF- ␬ B complexes in primary keratinocytes. Following incubation of primary keratinocytes with TRAIL (250 ng/ml) for 4 h, supershift electrophoretic mobility shift analysis of inducible NF- ␬ B-speci fi c complexes was performed with nuclear extracts. Each of the Rel-speci fi c antibodies (1 ␮ l of antibody [Ab]; see the text) was added to nuclear protein extracts. Control supershift electrophoretic mobility shift assays (EMSAs) were performed with normal rabbit serum (not shown). Arrows indicate supershifted complexes. (D) MG115 leads to dose-dependent inhibition of TRAIL-induced NF- ␬ B activation (upper panel) and I ␬ B ␣ degradation (lower panel). Membranes were rehybridized with antitubulin monoclonal antibodies to con fi rm comparable loading of cytoplasmic proteins.
… 
Caspases 8 and 10 are recruited and cleaved at the native TRAIL DISC independent of MG115. (A) Primary keratinocytes were treated with 10 ␮ M MG115 or diluent for 6 h and subsequently ana- lyzed for surface expression of TRAIL-R1 and TRAIL-R2 by fl uores- cence-activated cell sorting. MG115 does not modulate TRAIL-R1 or TRAIL-R2 surface expression on primary keratinocytes. (B) Analysis of the native TRAIL DISC in primary keratinocytes. Cells were treated for the indicated times with MG115 or diluent alone. The stimulated TRAIL DISC ( ϩ ) or unstimulated receptors ( Ϫ ) were precipitated with Flag-TRAIL precomplexed with 2 ␮ g of anti-Flag antibodies per ml (clone M2). The resulting protein complexes were separated by SDS-PAGE and analyzed by Western blot for compo- nents of the TRAIL DISC. The adaptor protein FADD was detectable only when stimulated receptors were precipitated. Also, recruitment of the two isoforms of caspase 8, caspase 8a and 8b, was detected only following stimulation of TRAIL receptors. In addition, the intermedi- ate cleavage products, p43/p41, of the two isoforms of caspase 8 were detectable in the DISC. For caspase 10, the full-length forms caspase 10d (p59) and caspase 10a (p55) as well as the cleavage products p47/43 and the p25 fragment, corresponding to the prodomain (Pro- dom.), were detectable in the immunoprecipitates. Analysis of cFLIP levels revealed that both isoforms, cFLIP L and cFLIP S , were present in the TRAIL DISC, with comparable levels in control cells and MG115- treated cells. For cFLIP L , only the p43 cleavage product was detectable in the DISC, indicative of proteolytic processing by active caspases in the DISC. The migration position of the IgG heavy and light chains of the precipitating anti-Flag M2 antibody as well as that of protein G cross-reacting with the antibody used for Western blot detection (caspase 10, lower part) are indicated by asterisks.
… 
Content may be subject to copyright.
MOLECULAR AND CELLULAR BIOLOGY, Feb. 2003, p. 777–790 Vol. 23, No. 3
0270-7306/03/$08.000 DOI: 10.1128/MCB.23.3.777–790.2003
Copyright © 2003, American Society for Microbiology. All Rights Reserved.
Proteasome Inhibition Results in TRAIL Sensitization of Primary
Keratinocytes by Removing the Resistance-Mediating Block
of Effector Caspase Maturation†
Martin Leverkus,
1
* Martin R. Sprick,
2
Tina Wachter,
1
Thilo Mengling,
1
Bernd Baumann,
3
Edgar Serfling,
4
Eva-B. Bro¨cker,
1
Matthias Goebeler,
1
Manfred Neumann,
4
and Henning Walczak
2
Department of Dermatology
1
and Institute of Pathology,
4
University of Wu¨rzburg Medical School, 97080 Wu¨rzburg,
Division of Apoptosis Regulation, Tumor Immunology Program, German Cancer Research Center, 69120
Heidelberg,
2
and Department of Physiological Chemistry, University of Ulm,
89081 Ulm,
3
Germany
Received 22 May 2002/Returned for modification 18 July 2002/Accepted 6 November 2002
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) exerts potent cytotoxic activity against
transformed keratinocytes, whereas primary keratinocytes are relatively resistant. In several cell types, inhi-
bition of the proteasome sensitizes for TRAIL-induced apoptosis by interference with NF-B activation. Here
we describe a novel intracellular mechanism of TRAIL resistance in primary cells and how this resistance is
removed by proteasome inhibitors independent of NF-B in primary human keratinocytes. This sensitization
was not mediated at the receptor-proximal level of TRAIL DISC formation or caspase 8 activation but further
downstream. Activation of caspase 3 was critical, as it only occurred when mitochondrial apoptotic pathways
were activated, as reflected by Smac/DIABLO, HtrA2, and cytochrome c release. Smac/DIABLO and HtrA2 are
needed to release the X-linked inhibitor-of-apoptosis protein (XIAP)-mediated block of full caspase 3 matu-
ration. XIAP can effectively block caspase 3 maturation and, intriguingly, is highly expressed in primary but
not in transformed keratinocytes. Ectopic XIAP expression in transformed keratinocytes resulted in increased
resistance to TRAIL. Our data suggest that breaking of this resistance via proteasome inhibitors, which are
potential anticancer drugs, may sensitize certain primary cells to TRAIL-induced apoptosis and could thereby
complicate the clinical applicability of a combination of TRAIL receptor agonists with proteasome inhibitors.
Apoptotic cell death is an important biological process that
is required to maintain the integrity and homeostasis of mul-
ticellular organisms. Inappropriate or impaired apoptosis has
been implicated in the development of many human diseases,
including cancer (71). The death-inducing members of the
tumor necrosis factor (TNF) family, TNF, CD95/APO-1/Fas
ligand (CD95L), and TNF-related apoptosis-inducing ligand
(TRAIL/APO-2L), have been studied most intensively. These
studies have resulted in elucidation of their role in activation-
induced cell death, autoimmune disorders, immune privilege,
and tumor evasion from the immune system (reviewed in ref-
erences 79, 82, and 85).
TRAIL has attracted attention for its ability to preferentially
kill tumor cells while most normal cells were resistant both in
vitro (56, 86) and in vivo (1, 20, 80). The functional analysis of
the TRAIL receptor-ligand system has been complicated by
the fact that a total of five different receptors for this cytokine
has been identified (reviewed in reference 46). Although the
major role of TRAIL is the induction of apoptosis, it has also
been shown in earlier overexpression studies to activate other
signaling pathways, such as the transcription factor NF-B (11,
62). NF-B is known to induce genes involved in apoptosis
resistance. Inhibition of NF-B can sensitize cells for TNF-or
TRAIL-induced apoptosis, depending on the cell type, sug-
gesting that distinct signaling pathways modulate the effect of
TRAIL in a cell type-specific manner (2, 27, 76).
The early biochemical events resulting in apoptosis induc-
tion by ligand-induced death receptor cross-linking have been
studied by the analysis of the so-called death-inducing signal-
ing complex (DISC) (33, 81). Cross-linking of CD95 or the two
apoptosis-inducing TRAIL receptors (TRAIL-R1 and
TRAIL-R2) results in the recruitment of Fas-associated death
domain (FADD; also called MORT1) and caspase 8 to the
DISC (3, 34, 68). In a homotypic interaction, the death domain
of FADD binds to the death domain of CD95. The death
effector domain of FADD in turn interacts with the death
effector domain of procaspase 8 and thereby recruits this
proenzyme to the DISC (51). Procaspase 8 is proteolytically
cleaved and thereby activated at the DISC. Activated caspase
8 then initiates the apoptosis-executing caspase cascade (81).
This cascade is further controlled by “cross talk” between the
intrinsic (mitochondrial) and extrinsic (death receptor) cell
death pathways, thereby modulating the outcome of death
receptor triggering (58). In that respect it has been shown for
CD95L and recently also for TRAIL (24, 41) that its proapo-
ptotic signaling can be blocked by Bcl-2 or Bcl-X
L
overexpres
-
sion in some cell types, whereas other cell types cannot be
protected by overexpression of these molecules, leading to the
* Corresponding author. Mailing address: Department of Derma-
tology, University of Wu¨rzburg Medical School, Josef-Schneider-Str. 2,
97080 Wu¨rzburg, Germany. Phone: 49 931 201 26356. Fax: 49 931 201
26700. E-mail: leverkus_m@klinik.uni-wuerzburg.de.
† This article is dedicated to Harald zur Hansen on the occasion of
his retirement as head of the German Cancer Research Center (Deut-
sches Krebs forschungszentrum) in Heidelberg with gratitude and ap-
preciation for 20 years of leadership.
777
concept of two different cell types utilizing distinct signaling
pathways with or without the necessity for mitochondrial con-
tribution (60).
Further complexity is added to the regulatory pathways in-
volved in death receptor sensitivity by proteins that are capable
of inhibiting active caspases. These proteins are called inhibi-
tor-of-apoptosis proteins (IAPs) (14, 25). IAPs are a family of
proteins dened by baculovirus repeat (BIR) domains and, in
some cases, a zinc ring nger domain (14, 25). IAPs like X-
linked IAP (XIAP), Livin/MLIAP, cIAP1, and cIAP2 block
apoptosis by directly inhibiting caspases. For some IAPs, an
involvement in caspase-independent pathways of apoptosis was
postulated (14, 65). XIAP is the most potent inhibitor of
caspases among the above-mentioned IAPs (14).
The ubiquitin-proteasome pathway plays a central role in
the regulation of essential cellular processes such as cell cycle
control, transcription, signal transduction, and apoptosis (28,
36). Many key regulatory proteins are controlled by ubiquiti-
nation, which targets them for degradation by the 26S protea-
some (29, 39). A recent report suggested that proteasome
inhibitors are able to induce apoptosis in transformed but not
in normal lymphocytes (50). Well-known proteasomal targets
include the NF-B/IB system, p53, and IAPs (28). Earlier
studies showed that NF-B activation is efciently blocked by
proteasome inhibitors (27). These data support the view that
sensitization to TNF- or TRAIL-induced apoptosis by pro-
teasome inhibitors is explained by the inhibition of NF-B
transcriptional activity (27, 30).
The mechanisms responsible for the resistance of primary
cells to TRAIL are not fully understood. Although initial re-
ports suggested that differential expression of TRAIL-R3 and
TRAIL-R4 might determine resistance to TRAIL in primary
cells, later studies demonstrated the important contribution of
intracellular regulatory pathways in primary cells (42). To fur-
ther investigate the resistance to TRAIL in primary human
keratinocytes, we analyzed proteasome-mediated resistance to
TRAIL-induced apoptosis and its neutralization by protea-
some inhibitors. We show that inhibition of proteasomal func-
tion effectively sensitizes primary keratinocytes to TRAIL in-
dependent of NF-B and downstream of the TRAIL DISC.
This sensitization required the activation of mitochondrial sig-
naling pathways necessary for XIAP inhibition. Our data sug-
gest that one important step to overcome the intracellular
resistance of primary keratinocytes to TRAIL is the inhibition
of XIAP by activation of mitochondrial apoptosis signaling
pathways.
MATERIALS AND METHODS
Materials. The protease inhibitor z-Val-Ala-Asp-uoromethyl ketone (zVAD-
FMK) was obtained from Bachem (Heidelberg, Germany). If not indicated
otherwise, all other reagents were of reagent grade and obtained from Serva
(Heidelberg, Germany). The following antibodies were used: poly(ADP-ribose)
polymerase (G. Poirier, CHUL Research Center, Quebec, Canada), caspase 3
(MF 393 and MF 397), and XIAP (MF 478; gifts from D. Nicholson, Merck
Frosst Corp, Quebec, Canada); Smac (kindly provided by M. MacFarlane, Le-
icester, United Kingdom); and HtrA2/Omi (generously provided by R. Taka-
hashi, RIKEN Brain Science Institute, Saitama, Japan). The FLICE (C-15) (51)
and cFLIP (NF-6) (61) monoclonal antibodies were kindly provided by P. H.
Krammer and are available from Alexis (Gruenberg, Germany). Monoclonal
antibodies against FADD and XIAP were purchased from Transduction Labo-
ratories (San Diego, Calif.), caspase 10 monoclonal antibodies (clone 4C1) were
from MBL International (Watertown, Mass.), cytochrome c monoclonal anti-
bodies were from Pharmingen (San Diego, Calif.), antibodies to IB (C-21)
were from Santa Cruz Biotechnology Inc. (Santa Cruz, Calif.), and antibodies to
IKK2 (antibody 2684) were from New England Biolabs, Boston, Mass.
The monoclonal antibodies specic for the different TRAIL receptors were
generated by immunizing mice with TRAIL receptor-Fc fusion proteins. The
specicity of the monoclonal antibodies was conrmed by staining cells trans-
fected with expression plasmids for TRAIL-R1 to TRAIL-R4 (H. Walczak,
unpublished observation). Anti-TRAIL-R1 (clone HS101) and anti-TRAIL-R2
(clone HS201), both of the murine immunoglobulin G1 (IgG1) isotype, were
used for uorescence-activated cell sorting staining. The monoclonal antibodies
are also available from Alexis (Gruenberg, Germany). Recombinant leucine
zipper (LZ)-TRAIL was generated as described before (78), and Flag-TRAIL
was expressed as previously described (32). Horseradish peroxidase-tagged don-
key anti-rabbit immunoglobulin antibodies and horseradish peroxidase-tagged
goat anti-mouse IgG were from Pharmingen (Hamburg, Germany), and horse-
radish peroxidase-conjugated goat anti-mouse IgG1 and IgG2b were obtained
from Southern Biotechnology Associates (Birmingham, Ala.).
Tissue culture. Primary keratinocytes were prepared from neonatal foreskins
as previously described (42). Cells were kept in serum-free KGM medium (Cell-
systems, St. Katharinen, Germany). Cells were only used up to passage 3 for all
experiments. The spontaneously transformed keratinocyte line HaCaT was
kindly provided by N. Fusenig (German Cancer Research Center, Heidelberg,
Germany) and cultured as described before (4).
Subfractionation of cytoplasmic proteins. Cytosolic extracts were generated
with a modied digitonin lysis protocol (66). Following trypsinization, cells were
washed with phosphate-buffered saline, permeabilized with digitonin (Fluka,
Taufkirchen, Germany) at a concentration of 150 g/ml for 10 min, and centri-
fuged at 5,000 g for 5 min to remove cellular debris. The resulting supernatants
were spun again at 13,000 g for 30 min at 4°C. Equal amounts of supernatant
were analyzed by Western blot. Blots were subsequently rehybridized with mono-
clonal antibodies to cytochrome c oxidase subunit IV (Molecular Probes Inc.,
Eugene, Oreg.) to control for mitochondrial lysis (data not shown).
Western blot analysis. Total cellular proteins were collected as described
before (44) with the exception that complete protease inhibitor cocktail (Roche
Molecular Diagnostics, Mannheim, Germany) was used. From 20 to 75 gof
protein was electrophoresed on sodium dodecyl sulfate-polyacrylamide gel elec-
trophoresis (SDS-PAGE) gels and transferred to nitrocellulose membranes.
Blocking of membranes and incubation with the indicated primary and appro-
priate secondary antibodies were performed essentially as described elsewhere
(43). Bands were visualized with the ECL detection kit (Amersham).
Analysis of death-inducing signaling complex. For precipitation of the native
TRAIL DISC, 5 10
6
human primary keratinocytes were used for each condi
-
tion. Cells were washed once with RPMI medium at 37°C and subsequently
incubated for the indicated time periods at 37°C in the presence of 1 gof
Flag-TRAIL per ml precomplexed with 2 g of anti-Flag antibody M2 (Sigma,
Taufkirchen, Germany) per ml for 15 min or, for the unstimulated control, in the
absence of Flag-TRAIL. DISC formation was stopped by washing the monolayer
twice with ice-cold phosphate-buffered saline. Cells were lysed on ice by addition
of 1 ml of lysis buffer (30 mM Tris-HCl [pH 7.5] at 21°C, 120 mM NaCl, 10%
glycerol, 1% Triton X-100, complete protease inhibitor cocktail [Roche Molec-
ular Diagnostics, Mannheim, Germany]). After 15 min of lysis, the lysates were
centrifuged at 50,000 g for 15 min to pellet cellular debris. DISC complexes
were precipitated from the lysates by coincubation with 20 l of protein G beads
(Roche, Mannheim, Germany) for 12 h on an end-over-end shaker at 4°C.
For precipitation of the nonstimulated receptors, 200 ng of Flag-TRAIL and
400 ng of anti-Flag M2 were added to the lysates prepared from nonstimulated
cells at 200 ng to control for protein association with a nonstimulated recep-
tor(s). Ligand afnity precipitates were washed ve times with lysis buffer before
the protein complexes were eluted from the beads by addition of 15 lof2
standard reducing sample buffer. Subsequently, proteins were separated by SDS-
PAGE on 4 to 12% NuPage Bis-Tris gradient gels (Invitrogen, Karlsruhe, Ger-
many) before detection of DISC components by Western blot analysis.
Determination of DEVDase activity. Cells were lysed following treatment in
buffer containing 50 mM HEPES-KOH (pH 7.0), 2 mM EDTA, 10% sucrose,
0.1% 3-[(3-cholamidopropyl)-dimethylammonio]-1-propanesulfonate (CHAPS),
and 5 mM dithiothreitol. Equal amounts of protein were incubated with 2,5 M
biotinylated DEVD-acyloxymethyl ketone (biotin-DEVD-AOMK) (kindly pro-
vided by D. Nicholson, Merck Frosst Corp, Quebec, Canada) for 30 min at 37°C
(57). Biotin-DEVD-AOMK is an irreversible caspase inhibitor that covalently
binds to DEVD-cleaving caspases, resulting in the biotin labeling of active
caspase fragments (57, 72). Samples were separated by SDS-PAGE and trans-
ferred to nitrocellulose membranes. Following overnight incubation with perox-
idase-conjugated streptavidin (Dako Diagnostika, Hamburg, Germany), biotin-
778 LEVERKUS ET AL. MOL.CELL.BIOL.
labeled fragments were visualized with the ECL detection kit (Amersham,
Arlington Heights, Ill.). Membranes were subsequently stripped and reprobed
with antibodies to caspase 3 (MF 393) to detect caspase 3-specic fragments.
Coimmunoprecipitation of XIAP. Primary and transformed keratinocytes (2
10
6
cells) were treated with 250 ng of TRAIL per ml for 3 h and lysed in TENT
buffer (50 mM Tris [pH 8.0], 2 mM EDTA, 150 mM NaCl, and 1% Triton X-100)
on ice. Equal amounts of cellular proteins were precipitated with 1 g of either
rabbit IgG, rabbit caspase 3 (MF 397), or XIAP (MF 478) antiserum (kindly
provided by D. Nicholson, Merck Frosst Corp, Quebec, Canada) in the presence
of 20 l of protein A-Sepharose beads overnight. Precipitates were washed four
times with lysis buffer before the protein complexes were eluted from the beads
by addition of 2 standard reducing sample buffer. Subsequently, proteins were
separated by SDS-PAGE before detection of XIAP by Western blot analysis with
monoclonal antibodies to XIAP (Transduction Laboratories, San Diego, Calif.).
FACScan analysis. For analysis of the mitochondrial transmembrane potential
(⌬⌿m), 5 10
5
cells were resuspended in Dulbeccos modied Eagles medium
containing 40 nM tetramethylrhodamine ethyl ester (59) and incubated at 37°C
for 15 min as described before (21). After being washed once with ice-cold
phosphate-buffered saline, cells were immediately analyzed. For surface staining
of TRAIL-R1 and TRAIL-R2, 2 10
5
cells were detached by incubation with
phosphate-buffered saline containing 5 mM EDTA, followed by brief trypsiniza-
tion, and incubated with monoclonal antibodies against TRAIL-R1 (HS101),
TRAIL-R2 (HS201), or control murine IgG1 at 10 g/ml, followed by biotinyl-
ated secondary goat anti-mouse immunoglobulin antibodies and phycoerythrin-
conjugated streptavidin (Pharmingen, Hamburg, Germany). For all experiments,
10
4
cells were analyzed by FACScan (Becton Dickinson & Co, San Jose, Calif.).
Apoptosis and cytotoxicity assays. Crystal violet staining of surviving attached
cells was performed 16 to 24 h after addition of LZ-TRAIL in 24-well plates (1
10
5
cells) or 96-well plates (2 10
4
cells) as described before (42). Parallel
plates were examined by propidium iodide staining and ow cytometric analysis
of subdiploid DNA content as described before (42). For annexin-propidium
iodide staining, 2 10
5
to 5 10
5
cells were stained with 10 l of annexin
V-uorescein isothiocyanate conjugate (Becton Dickinson & Co, San Jose, Cal-
if.) and 5 l of propidium iodide (50 ng/ml) according to the manufacturers
instructions, incubated for 15 min at room temperature in the dark, and imme-
diately analyzed.
Electrophoretic mobility shift assays. Nuclear and cytoplasmic extracts of
primary keratinocytes were prepared according to Schreiber et al. (63) with slight
modications as previously described (53). The oligonucleotide probe used was
a consensus NF-B binding site derived from the B element of the murinein-
terleukin-2 promoter (TCEd
AC
) (6): 3-GACCAAGAGGGATTTCACCCCT
AAATC-5. The following antibodies were used for the supershift electro-
phoretic mobility shift assay: anti-p65-NF-B (Santa Cruz Biotechnology Inc.,
Santa Cruz, Calif.; A, sc-109), anti-c-Rel (Santa Cruz; N, sc-070), anti-RelB
(Santa Cruz; C-19, sc-226), anti-p50-NF-B (Santa Cruz; NLS, sc-114), and
anti-p52-NF-B (Upstate Biotechnology Inc., Lake Placid, N.Y.; 06-413).
Retroviral infection. The pCFG5-IEGZ retroviral vector containing inserts of
the transdominant ⌱␬B (⌱␬B-TD) mutant or kinase-dead IKK2 (IKK2-KD)
mutant was used for infection of primary human keratinocytes as described
earlier (13) with the exception that viral supernatants were generated in KGM.
Infection efciency of keratinocytes ranged from 30% to 80%, as determined by
green uorescent protein uorescence 72 h after infection. At this time point,
keratinocytes were used for cytotoxicity assays and biochemical characterization.
Transient-transfection assay for XIAP. Cells were transfected with the
Nucleofector technology (Amaxa, Ko¨ln, Germany) according to the recom-
mended protocols. Briey, 2 10
6
transformed keratinocytes were resuspended
in 100 l of Cell Line R solution. Then 3 g of pEBB-XIAP vector or pEBB
control vector (kindly provided by Colin Duckett, University of Michigan Med-
ical School) was mixed with the suspension, added to an Amaxa cuvette, and
nucleofected with program U27. Cells were then diluted with 500 l of warm
RPMI and added to six-well plates containing 1 ml of warm medium. After a 10-h
incubation period at 37°C, biochemical and functional studies were performed as
indicated in the gure legends. Transfection efciency, as determined by green
uorescent protein expression in parallel plates, varied between 50% and 70%.
RESULTS
Primary keratinocytes are sensitized to TRAIL-induced apo-
ptosis by proteasome inhibition. We have previously shown
that primary keratinocytes are relatively resistant to TRAIL-
induced apoptosis compared to transformed keratinocytes
(42). Inhibition of proteasome function modulates apoptotic
signaling pathways in a variety of tumor cells (16, 28). We thus
explored the potential role of proteasomal function for the
resistance of primary keratinocytes to TRAIL-mediated apo-
ptosis. To this end, we used MG115 as a proteasomal inhibitor
in our studies. MG115 treatment resulted in a dramatic sensi-
tization of primary keratinocytes for TRAIL-induced apoptosis
in a concentration-dependent manner (Fig. 1) (55). Moreover,
two other known proteasome inhibitors, lactacystin (19) and
epoxomicin (52), similarly sensitized primary keratinocytes for
TRAIL-induced apoptosis (data not shown). The broad-spec-
trum caspase inhibitor zVAD-FMK completely abolished
TRAIL-induced apoptosis under these conditions, conrming
the critical role of caspases in this process (Fig. 1). Our data
suggest that peptide aldehyde inhibitors such as MG115 inter-
fere with proteasome-regulated signaling pathways, resulting
FIG. 1. Proteasome inhibitor MG115 sensitizes primary keratino-
cytes to TRAIL-induced apoptosis. (A) Primary keratinocytes were
treated with 50 ng of TRAIL per ml for 16 h. Caspase-dependent
cytotoxicity of TRAIL (sub-G
0
DNA analysis) is only detectable in the
presence of the proteasome inhibitor MG115 (10 M). zVAD-FMK
(40 M) completely abolishes TRAIL-induced apoptosis. (B) Dose-
dependent effect of MG115 on TRAIL-induced cell death in primary
keratinocytes was determined by crystal violet staining 16 h after ad-
dition of the indicated concentrations of TRAIL.
V
OL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 779
in increased sensitivity of primary keratinocytes to TRAIL-
induced apoptosis.
TRAIL induces NF-B activation in primary keratinocytes.
One important target of proteasomal degradation is the IB/
NF-B system (29, 48). NF-B has potent antiapoptotic prop-
erties, particularly in TNF-induced apoptosis (30), whereas
its role in TRAIL-induced apoptosis is more controversial (26,
27). To study whether the increased sensitivity to TRAIL-
induced apoptosis may be mediated by inhibition of NF-B
function, we rst analyzed TRAIL-induced NF-B activation
in primary keratinocytes and the impact of the proteasome
inhibitor MG115. Employing electrophoretic mobility shift as-
says, we detected an increase in NF-B DNA binding activity
upon stimulation with TRAIL at concentrations of between 50
and 500 ng/ml (Fig. 2A). This induction was observed within 30
min and increased further up to 6 h after stimulation (Fig. 2B).
Although zVAD-FMK has been shown to completely block
TRAIL-induced apoptosis (42), it did not negatively affect
TRAIL-induced NF-B activation (Fig. 2B), indicating that
caspase activation is not required for TRAIL-induced NF-B
activation. The supershift electrophoretic mobility shift assay
demonstrated that TRAIL-induced NF-B binding activity
consisted mainly of p50 homodimers as well as p65/p50 het-
erodimers. Nevertheless, weak supershift signals of p65/c-Rel
complexes were also detectable (Fig. 2C). In line with reports
for other cell types (27), TRAIL-induced NF-B DNA binding
activity and degradation of IB were dose-dependently inhib-
ited by the proteasome inhibitor MG115 (Fig. 2D).
NF-B activation does not modulate TRAIL sensitivity in
keratinocytes. The proteasome has been shown to modulate
multiple signaling pathways, including NF-B, p53, and IAPs
(9, 88). In order to inhibit NF-B more specically, we next
studied whether ectopic expression of dominant negative mu-
tants of IKK2 (IKK2-KD) or ⌱␬B (⌱␬B-TD) inuenced
FIG. 2. TRAIL dose-dependently enhances NF-B binding in primary keratinocytes. (A) Primary keratinocytes were treated with 5 to 1,000
ng of TRAIL per ml for 6 h and analyzed for NF-B DNA binding by electrophoretic mobility shift assay with nuclear extracts. Induction of p65/p50
heterodimers (upper arrow) or p50 homodimers (lower arrow) is shown. As a control for the specicity of DNA binding, control experiments with
an excess of unlabeled probe were performed (not shown). Cell viability was determined in parallel experiments by crystal violet staining and is
indicated as a percentage of the control value. (B) Primary keratinocytes were exposed to 250 ng of TRAIL per ml for6hinthepresence or
absence of 40 M zVAD-FMK. Survival of cells was determined in parallel experiments as described for A. (C) Composition of TRAIL-induced
nuclear NF-B complexes in primary keratinocytes. Following incubation of primary keratinocytes with TRAIL (250 ng/ml) for 4 h, supershift
electrophoretic mobility shift analysis of inducible NF-B-specic complexes was performed with nuclear extracts. Each of the Rel-specic
antibodies (1 l of antibody [Ab]; see the text) was added to nuclear protein extracts. Control supershift electrophoretic mobility shift assays
(EMSAs) were performed with normal rabbit serum (not shown). Arrows indicate supershifted complexes. (D) MG115 leads to dose-dependent
inhibition of TRAIL-induced NF-B activation (upper panel) and IB degradation (lower panel). Membranes were rehybridized with antitubulin
monoclonal antibodies to conrm comparable loading of cytoplasmic proteins.
780 LEVERKUS ET AL. M
OL.CELL.BIOL.
TRAIL sensitivity in keratinocytes. We thus examined primary
keratinocytes following retroviral infection with either IKK2-
KD, ⌱␬B-TD, or a viral control construct. Similar to the data
shown in Fig. 2D, overexpression of these proteins efciently
inhibited TRAIL-induced degradation of ⌱␬B and NF-B
activation compared to vector-transfected control cells (Fig.
3A and data not shown). However, sensitivity to TRAIL-in-
duced apoptosis was not signicantly modulated by these mu-
tants (Fig. 3B).
Consistent with these data, neither TRAIL nor the protea-
some inhibitor MG115 modulated mRNA levels of antiapo-
ptosis genes such as cIAP-1, cIAP-2, TRAF-1, or TRAF-2 in
keratinocytes when assayed by an RNase protection assay (not
shown). Taken together, our results indicate that neither
TRAIL-induced NF-B activation nor constitutive NF-B ac-
tivation signicantly modulates resistance to TRAIL. More
importantly, our data suggest that the peptide aldehyde inhib-
itor MG115 interferes with other proteasome-regulated signal-
ing pathways which nally result in increased sensitivity of
keratinocytes to TRAIL-induced apoptosis.
Sensitization to TRAIL-induced apoptosis by proteasome
inhibition is mediated downstream of the death-inducing sig-
naling complex. We next determined whether proteasome in-
hibitors modulate surface expression of the proapoptotic
TRAIL-R1 and TRAIL-R2. As shown in Fig. 4A, exposure of
primary keratinocytes to MG115 did not signicantly modulate
surface expression of either TRAIL-R1 or TRAIL-R2, which
are both expressed in primary keratinocytes. TRAIL stimula-
tion of these receptors leads to rapid activation of caspase 8
and, as recently shown, also of caspase 10 (35, 67, 84) at the
death-inducing signaling complex (DISC) initiating the
caspase cascade (3, 34, 68). Thus, increased caspase 8 or 10
recruitment to and/or subsequent activation at the DISC could
be responsible for the observed sensitization to TRAIL-in-
duced apoptosis following MG115 treatment.
We therefore examined whether MG115 modies the acti-
vation of caspase 8 or 10 at the DISC by immunoprecipitation
of the native TRAIL DISC from primary keratinocytes (Fig.
4B). DISC analysis demonstrated rapid recruitment and cleav-
age of caspases 8 and 10 at the DISC. The structurally related
protein cFLIP, which is also expressed in primary keratino-
cytes, acts as an inhibitor of CD95L- and TRAIL-induced
apoptosis by inhibiting the activation of caspase 8 at the DISC
(38, 42, 61). In line with our previous observation (42), large
amounts of cFLIP
L
and cFLIP
S
(where L indicates long and S
indicates short) were found in the DISC. Importantly, the
DISC composition remained unchanged after treatment with
the proteasome inhibitor MG115 (Fig. 4B). Thus, neither
DISC activation nor surface expression of the TRAIL-R pro-
teins explains the increased sensitivity to TRAIL of primary
keratinocytes treated with MG115. However, it is currently not
known whether recruitment and activation of caspases is the
sole predictor of the activity of DISC constituents. Therefore,
we next examined activation kinetics of the known DISC-as-
sociated proteins in cellular lysates of primary keratinocytes in
the presence and absence of MG115.
Indeed, TRAIL induced rapid cleavage of caspase 8 to
p43/41 as well as to the active p18 fragment, which were de-
tectable within 60 to 120 min of stimulation. Furthermore,
rapid cleavage of cFLIP
L
(p43) was seen within 60 to 120 min.
Similarly, specic cleavage of caspase 10 to the p47/p43 inter-
mediates as well as to a 25-kDa fragment representing the fully
cleaved prodomain was detectable (Fig. 5). Slight differences in
the amount of the active fragment of caspase 8 (p18) or the
fully cleaved prodomain of caspase 10 (p25) were observed at
later time points in the presence of the inhibitor MG115.
Addition of zVAD-FMK fully inhibited TRAIL-induced cleav-
age of caspase 8, caspase 10, and, to a lesser extent, cFLIP
L
(Fig. 5). In the presence of MG115, levels of cFLIP
S
were
increased compared to the barely detectable levels found un-
der control conditions.
These data show that MG115-induced sensitization of pri-
mary keratinocytes to TRAIL is not regulated at the level of
caspase 8 or 10 activation. Collectively, our ndings indicate
FIG. 3. Overexpression of dominant negative mutant of IKK2
(IKK2-KD) or transdominant IB (IB-TD) inhibits TRAIL-in-
duced degradation of IB without modulation of TRAIL-induced
apoptosis. (A) Primary human keratinocytes were retrovirally trans-
duced with IKK2-KD, IB-TD, or control vector and subsequently
analyzed for expression of mutant proteins as well as degradation of
endogenous IB following stimulation with 500 ng of TRAIL per ml
for1hbyWestern blotting. Membranes were rehybridized with anti-
tubulin monoclonal antibodies to conrm comparable loading of cel-
lular proteins. (B) Primary keratinocytes overexpressing IKK2-KD or
IB-TD as described for A were either left untreated or incubated
with 25, 50, or 125 ng of LZ-TRAIL per ml for 6 h. Thereafter, the
percentage of dead green uorescent protein-positive cells was deter-
mined by tetramethylrhodamine ethyl ester staining. Data are shown
as mean standard deviation of two independent experiments.
V
OL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 781
that the dramatic sensitization of primary keratinocytes for
TRAIL-mediated apoptosis induced by proteasome inhibition
is not due to differential activation of the apoptotic signaling
pathway at the TRAIL DISC. Thus, this sensitization of pri-
mary keratinocytes for TRAIL-induced apoptosis must be reg-
ulated at a step downstream of the DISC.
Autocatalytic maturation of caspase 3 to active enzyme is
blocked in primary keratinocytes. Caspase 3 is the major ef-
fector caspase in most cell types (54). The activation of caspase
3 is tightly regulated at several levels, beginning with a proteo-
lytic cut between the large and small subunits, followed by
removal of the prodomain. While the rst step occurs mainly
via active initiator caspases such as caspase 8, 9, or 10, the
second step is mediated by autoproteolytic maturation of
caspase 3 (54). In order to further examine the level at which
MG115 acts to sensitize primary keratinocytes for TRAIL-
induced apoptosis, we examined the activation pattern of
caspase 3. Interestingly, TRAIL stimulation of primary kera-
tinocytes in the absence of MG115 resulted in the cleavage of
caspase 3 to a 20-kDa fragment within 60 to 120 min without
detectable induction of apoptosis. However, full processing to
the 17-kDa fragment was detectable when cells were treated
with TRAIL in the presence of MG115 (Fig. 6A).
To further examine the activity of caspase 3 under these
conditions, we used biotinylation of catalytically active caspase
FIG. 4. Caspases 8 and 10 are recruited and cleaved at the native
TRAIL DISC independent of MG115. (A) Primary keratinocytes were
treated with 10 M MG115 or diluent for 6 h and subsequently ana-
lyzed for surface expression of TRAIL-R1 and TRAIL-R2 by uores-
cence-activated cell sorting. MG115 does not modulate TRAIL-R1 or
TRAIL-R2 surface expression on primary keratinocytes. (B) Analysis
of the native TRAIL DISC in primary keratinocytes. Cells were
treated for the indicated times with MG115 or diluent alone. The
stimulated TRAIL DISC () or unstimulated receptors () were
precipitated with Flag-TRAIL precomplexed with 2 g of anti-Flag
antibodies per ml (clone M2). The resulting protein complexes were
separated by SDS-PAGE and analyzed by Western blot for compo-
nents of the TRAIL DISC. The adaptor protein FADD was detectable
only when stimulated receptors were precipitated. Also, recruitment of
the two isoforms of caspase 8, caspase 8a and 8b, was detected only
following stimulation of TRAIL receptors. In addition, the intermedi-
ate cleavage products, p43/p41, of the two isoforms of caspase 8 were
detectable in the DISC. For caspase 10, the full-length forms caspase
10d (p59) and caspase 10a (p55) as well as the cleavage products
p47/43 and the p25 fragment, corresponding to the prodomain (Pro-
dom.), were detectable in the immunoprecipitates. Analysis of cFLIP
levels revealed that both isoforms, cFLIP
L
and cFLIP
S
, were present in
the TRAIL DISC, with comparable levels in control cells and MG115-
treated cells. For cFLIP
L
, only the p43 cleavage product was detectable
in the DISC, indicative of proteolytic processing by active caspases in
the DISC. The migration position of the IgG heavy and light chains of
the precipitating anti-Flag M2 antibody as well as that of protein G
cross-reacting with the antibody used for Western blot detection
(caspase 10, lower part) are indicated by asterisks.
FIG. 5. Proteasome inhibitor MG115 does not inuence early
TRAIL-induced caspase 8, caspase 10, and cFLIP cleavage. Primary
keratinocytes were incubated for 60 min with 10 M MG115 or diluent
alone and subsequently treated with 50 ng of TRAIL per ml for the
indicated times. TRAIL treatment led to cleavage of caspase 8 (p43/
p41/p18), caspase 10 (p47/43/25), and cFLIP (p43) under both condi-
tions, as determined by Western blot analysis. Membranes were rehy-
bridized with antitubulin monoclonal antibodies to conrm comparable
loading of proteins.
782 LEVERKUS ET AL. M
OL.CELL.BIOL.
3 in cellular lysates of primary keratinocytes (57). This tech-
nique allows the simultaneous detection of enzymatic activity
and molecular weight, enabling the parallel monitoring of cleav-
age and activity of caspase 3. Biotin-labeled fragments of
caspase 3 were clearly detectable in lysates of cells treated with
TRAIL in the presence of MG115. Interestingly, in cells
treated with TRAIL alone, full-length caspase 3 was marginally
labeled, indicating some enzymatic activity of the full-length
caspase 3, conrming the recent report by Roy et al. (57) (Fig.
6B, upper panel). Consistent with the data shown in Fig. 6A,
reprobing of the same membrane with caspase 3-specic anti-
bodies revealed the comparable presence of the cleaved p20
fragment of caspase 3 in lysates treated with TRAIL alone
(Fig. 6B, middle panel). These data demonstrate that the ini-
tial cleavage of caspase 3 is not blocked in TRAIL-treated
primary keratinocytes but that the resulting cleavage product
was enzymatically inactive. In line with this observation, cleav-
age of poly(ADP-ribose) polymerase, a known substrate of
caspase 3 (40), was only detected when caspase 3 was further
processed to the p17 fragment after treatment with MG115
(Fig. 6B, lower panel). Taken together, our ndings indicate
that TRAIL-induced full processing of caspase 3 to the active
17-kDa fragment which is required for full enzymatic activity is
blocked in primary human keratinocytes and that this block is
released upon treatment of primary keratinocytes with the
proteasome inhibitor MG115.
Differential expression of XIAP in primary versus trans-
formed keratinocytes. In order to further investigate the inter-
play between caspase 3 maturation and TRAIL receptor trig-
gering, we compared two parameters of TRAIL-induced
apoptosis in primary versus transformed keratinocytes: the
dose-dependent cleavage pattern of caspase 3 and induction of
apoptosis as determined by hypodiploidy analysis (Fig. 7). In
line with our previous ndings (42), low concentrations of
TRAIL were sufcient to induce signicant apoptosis in trans-
formed keratinocytes, whereas primary keratinocytes were
dose-dependently protected but nonetheless succumbed to
high concentrations of TRAIL (Fig. 7). In contrast to the
ndings in primary keratinocytes (Fig. 7, lower panel, left
lanes), the p20 fragment of caspase 3 was hardly detectable in
transformed keratinocytes, whereas the p17 and a p15 frag-
ment of caspase 3 were readily detected in these cells when
stimulated with low concentrations of TRAIL (Fig. 7, lower
panel, right lanes). Full cleavage correlated with detectable
activity of effector caspases, as examined by afnity labeling
(Fig. 7, upper panel). Interestingly, high concentrations of
TRAIL sufcient to induce partial apoptosis led to labeling of
full-length caspase 3 in primary keratinocytes (Fig. 7, upper
panel).
Taken together, these data pointed to a potential inhibitor
of effector caspases blocking the full maturation of cleaved
caspase 3 in primary keratinocytes. XIAP, the most potent
caspase inhibitor of the IAP family, has been shown to selec-
tively inhibit the cleaved forms of caspase 3 and 7 (14). We
therefore examined the expression level of XIAP in primary
and transformed keratinocytes. Intriguingly, Western blot
analysis revealed that the 57-kDa XIAP protein was present in
primary but not in transformed keratinocytes (Fig. 8A, lysate).
Furthermore, a 29-kDa fragment of XIAP generated by
caspase-mediated cleavage (15) was only detected in TRAIL-
FIG. 6. Full processing of caspase 3 to a 17-kDa fragment in
TRAIL-treated primary keratinocytes upon proteasome inhibition.
(A) Primary keratinocytes were incubated for 60 min with 10 M
MG115 (right part) or diluent alone (left part) and subsequently
treated with 50 ng of TRAIL per ml for the indicated time intervals.
TRAIL leads to substantial cleavage of caspase 3 to a 20-kDa fragment
(p20), whereas only in the presence of the proteasome inhibitor is a
17-kDa fragment (p17) detectable. No cleavage fragments were de-
tected in the presence of zVAD-FMK or after incubation with MG115
alone. Membranes were rehybridized with antitubulin monoclonal an-
tibodies to conrm comparable loading of proteins. (B) TRAIL-in-
duced effector caspase activity with afnity labeling with biotinylated
DEVD-AOMK (see Materials and Methods). Activity of cleaved ef-
fector caspases by 50 ng of TRAIL per ml requires the presence of the
proteasome inhibitor MG115. Primary keratinocytes were treated as
above, and DEVD-specic caspase activity in cellular lysates was an-
alyzed 3 h after TRAIL stimulation. DEVDase activity of cleaved
fragments (p17/p15, upper panel) was only detectable in cells treated
with TRAIL in the presence of MG115, whereas TRAIL or MG115
alone did not induce detectable caspase activity. Slight labeling of
full-length caspase 3 was detected in cells treated with TRAIL alone.
Subsequent rehybridization of the blot with caspase 3-specic antibody
conrmed that the slower-migrating caspase 3 species (p20) did not
have detectable enzymatic activity in primary keratinocytes (middle
panel). In line, poly(ADP-ribose) polymerase (PARP) cleavage was
only detected upon treatment with TRAIL in the presence of MG115
(lower panel). Membranes were rehybridized with antitubulin mono-
clonal antibodies to conrm comparable loading of proteins.
V
OL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 783
treated primary keratinocytes. These data suggest that XIAP
may act as an inhibitor of TRAIL-induced apoptosis in primary
keratinocytes by blocking the full cleavage and, thus, the ac-
tivity of caspase 3.
In order to investigate further the interaction of caspase 3
with XIAP, immunoprecipitation studies were performed.
While antibodies to XIAP readily precipitated XIAP in un-
treated as well as TRAIL-treated primary keratinocytes, coim-
munoprecipitation of XIAP with caspase 3 was only detected
following TRAIL treatment (Fig. 8A). These data demonstrate
the interaction of caspase 3 with XIAP in TRAIL-treated pri-
mary keratinocytes under physiological conditions.
In order to address the question of whether the lack of XIAP
in transformed keratinocytes contributes to the high TRAIL
sensitivity of these cells, transient-transfection studies were
performed. Following overexpression of XIAP in transformed
keratinocytes (Fig. 8B, middle lanes) resistance to TRAIL-
induced apoptosis was increased (Fig. 8C). Furthermore, in-
creased levels of caspase 3 p32 and p20 were detected follow-
ing TRAIL treatment in XIAP-overexpressing cells.
Interestingly, high levels of overexpressed XIAP (Fig. 8B, up-
per panel) did not completely block TRAIL-induced caspase 3
maturation compared to primary keratinocytes (Fig. 8B, lower
panel). Because the transfection efciency in these experi-
ments was between 50 and 70%, the remaining detectable fully
cleaved caspase 3 (Fig. 8B) as well as the partial apoptosis
protection in functional assays (Fig. 8C) might, at least in part,
be explained by the percentage of cells without efcient XIAP
expression. However, we cannot exclude that additional differ-
ences in the inhibition of effector caspase activation exist be-
tween primary and transformed keratinocytes. Future studies
are required to delineate this point. Taken together, our data
demonstrate that the lack of XIAP contributes to the higher
sensitivity of transformed keratinocytes to TRAIL.
We next investigated whether the expression level or cleav-
age pattern of XIAP following TRAIL treatment of primary
keratinocytes is modulated by MG115. We therefore examined
XIAP levels during TRAIL treatment in the presence and
absence of MG115. Limited cleavage of XIAP to a 29-kDa
fragment was detected in the presence and absence of MG115.
However, a slight increase in cleaved XIAP was detected at
later time points in MG115-treated cells (Fig. 8D). This in-
crease correlated with full caspase 3 processing (compare Fig.
6A), suggesting that XIAP cleavage is a secondary event me-
diated by active caspase 3. Importantly, MG115 treatment
alone did not signicantly change XIAP levels (Fig. 8D). These
data provide evidence that the mechanism of MG115-induced
increase of TRAIL sensitivity does not involve cleavage of
XIAP or modulation of XIAP expression but rather direct
inhibition of caspase 3 by XIAP.
MG115 sensitizes primary keratinocytes to TRAIL-induced
apoptosis upstream of or at the mitochondrial level. Apoptotic
activation of the mitochondrion results in the release of pro-
teins such as cytochrome c, Smac/DIABLO, and HtrA2/Omi
from the intermitochondrial membrane space (49, 73). Smac/
DIABLO and HtrA2/Omi release from mitochondria in turn
leads to caspase activation by inhibiting the antiapoptotic func-
tion of several IAPs, particularly XIAP (8, 17, 18, 45, 74). Our
nding that XIAP is highly expressed in primary keratinocytes
prompted us to investigate whether proteasome inhibition may
allow TRAIL-induced release of Smac/DIABLO or HtrA2/
Omi from mitochondria, resulting in XIAP inhibition.
FIG. 7. Caspase 3 activation pattern in primary and transformed keratinocytes. (A) DEVD-specic caspase activity in cellular lysates was
analyzed 3 h after TRAIL stimulation in primary keratinocytes (PK) and transformed HaCaT keratinocytes (TK). DEVDase activity of p19, p17,
and p15 (upper panel) was detectable in transformed keratinocytes treated with TRAIL, whereas the same concentrations of TRAIL did not
induce detectable DEVDase activity of cleaved fragments of caspase 3 in primary keratinocytes. Induction of apoptosis was monitored in parallel
plates by hypodiploidy analysis. Higher concentrations of TRAIL sufcient to induce partial cell death in primary keratinocytes led to a
dose-dependent increase in full-length caspase 3 labeling, indicative of some enzymatic activity of full-length caspase 3. Subsequent reprobing of
blots with caspase 3-specic antibody conrmed that low concentrations of TRAIL were sufcient to induce the slower-migrating caspase 3 form
(p20) in primary keratinocytes. In contrast, mainly the mature cleaved forms p17 and a smaller fragment (p15) were detected in transformed
keratinocytes, even at low concentrations of TRAIL. Ponceau Red staining of the membrane conrmed comparable loading of proteins. HRP,
horseradish peroxidase.
784 LEVERKUS ET AL. M
OL.CELL.BIOL.
FIG. 8. XIAP confers resistance to TRAIL-induced apoptosis in primary keratinocytes. (A) XIAP is expressed in primary keratinocytes (PK)
but not in transformed keratinocytes (TK) and can be coimmunoprecipitated under native conditions with caspase 3 following TRAIL treatment.
Total cellular lysates from primary keratinocytes and transformed keratinocytes were prepared, and equal amounts of protein were either analyzed
by Western blotting (25 g) with XIAP monoclonal antibodies (lysate, left and right side) or subjected to immunoprecipitation (500 g). A 57-kDa
protein representing full-length XIAP was detected in primary (right) but not transformed (left) keratinocytes. An additional band of roughly 65
kDa (asterisks) detected in total lysates was shown to be a nonspecic band (immunoprecipitation: XIAP, right lanes). Following immunopre-
cipitation with caspase 3 antiserum, full-length but not cleaved XIAP was detected only in immunoprecipitates of TRAIL-treated primary
keratinocytes PK (immunoprecipitation: caspase 3). (B) Transfection of transformed keratinocytes with XIAP leads to decreased cleavage of
full-length caspase 3 and increased detection of caspase 3 p20. Transformed keratinocytes were transiently transfected with 3 g of pEBB-XIAP
vector or pEBB control vector as indicated in Materials and Methods. Then 125 ng of TRAIL per ml was added to the six-well plates, and cellular
lysates were collected 3 h later. Primary keratinocytes were treated in parallel plates. The same membrane was rst incubated with monoclonal
antibodies to XIAP and subsequently reprobed with caspase 3 antibodies. Rehybridization with antitubulin monoclonal antibodies conrmed
comparable loading of cellular proteins. Shown is a representative experiment of a total of three independent experiments. (C) XIAP overex-
pression leads to relative resistance of transformed keratinocytes to TRAIL-induced apoptosis. Cells were transfected as described for B and
treated with the indicated concentrations of TRAIL, and viability was determined 6 h later with annexin/propidium iodide staining. Shown are
means and standard deviations for two independent experiments. (D) Primary keratinocytes were incubated for 60 min with 10 M MG115 (right
part) or diluent alone (left part) and subsequently treated with 50 ng of TRAIL per ml for the indicated time periods. TRAIL treatment resulted
in cleavage of XIAP to a 29-kDa fragment (p29), with similar cleavage at early time points in the presence or absence of the proteasome inhibitor.
No cleavage fragments were detected in the presence of zVAD-FMK or after incubation with MG115 alone for 4 h, at which time XIAP levels
were comparable to those found under control conditions.
V
OL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 785
We therefore examined the kinetics of the release of mito-
chondrial proteins into the cytosol during MG115-induced sen-
sitization of primary keratinocytes to TRAIL. In cells treated
with TRAIL alone, release of cytochrome c, Smac/DIABLO,
or HtrA2/Omi could not be detected (Fig. 9A). In contrast, in
cells treated with TRAIL in the presence of MG115, cyto-
chrome c, Smac/DIABLO, and HtrA2/Omi were rapidly re-
leased from the mitochondria, whereas exposure to MG115
alone had no effect (Fig. 9A). Consistently, the quantitative
disruption of the inner mitochondrial membrane potential
(⌬␺
m
) which represents an important point of no return of
apoptosis (37), was detected in TRAIL-treated primary kera-
tinocytes in the presence but not in the absence of MG115
(Fig. 9B). Taken together, these data suggest that proteasome
inhibition sensitizes primary keratinocytes for TRAIL-induced
apoptosis by disabling the XIAP-mediated inhibition of auto-
proteolytic maturation of caspase 3 by allowing Smac/DIA-
BLO and HtrA2/Omi release from the mitochondria and, thus,
inhibition of XIAP.
DISCUSSION
Primary keratinocytes are resistant to TRAIL concentra-
tions that induce apoptosis in transformed keratinocytes but
nonetheless succumb to high levels of TRAIL (42). This ob-
servation led us to further explore intracellular signaling path-
ways in primary keratinocytes which might modulate sensitivity
to death ligands such as TRAIL (26, 27, 76). One important
step for the regulation of such signaling pathways is the pro-
grammed degradation of specic proteins via the proteasome
(16). Our initial nding that proteasome inhibition dramati-
cally sensitized primary cells to TRAIL led us to examine a
prime candidate for the regulation of TRAIL sensitivity, the
transcription factor NF-B. Several reports have suggested
that TNF-mediated apoptosis in particular is regulated by a
group of genes induced by NF-B, including cIAP-1, cIAP-2,
TRAF-1, and TRAF-2 (83, 87). Regarding TRAIL, conicting
data have been published (26, 27), suggesting that for this
death ligand, cell type-specic differences may exist. Here we
show that TRAIL induced a moderate but reproducible acti-
vation of NF-B-specic DNA binding and I␬␤␣ degradation
under nonoverexpression conditions of TRAIL-R proteins.
Our data are in line with recent reports utilizing 293 and HeLa
cell lines (23, 27, 76). However, specic blockade of NF-Bby
ectopically expressed inhibitors of NF-B activation (i.e., IKK2
or I␬␤␣) failed to sensitize keratinocytes to TRAIL. These
data clearly indicate that other signaling pathways modulated
by proteasome function play a more important role for sensi-
tivity to TRAIL, at least in keratinocytes.
TRAIL-induced signaling pathways have thus far been stud-
ied mainly in cell lines that are highly sensitive to TRAIL or in
cell lines that are resistant to receptor ligation due to articial
overexpression of intracellular inhibitors such as cFLIP (31).
In order to study the possible involvement of TRAIL DISC
formation in the mechanism of TRAIL resistance of primary
keratinocytes, we rst had to elucidate the composition of the
native TRAIL DISC in these cells. We determined that the
TRAIL DISC in primary keratinocytes contains FADD,
caspase 8, caspase 10, the second initiator caspase recently
shown to be recruited to the native TRAIL DISC in several cell
lines (35, 67, 84), cFLIP
L
, and cFLIP
S
. Earlier overexpression
studies have demonstrated that cFLIP
L
is capable of inhibiting
TRAIL-induced apoptosis in keratinocytes (42) and melanoma
cells (22). Detailed comparative and kinetic studies of the
proteolytic activity of the TRAIL DISC are required to deter-
mine the exact contribution of all DISC components in these
cells. Nonetheless, our studies clearly showed that in primary
keratinocytes, a cell type that expresses TRAIL-R1 and
TRAIL-R2 at high levels, inhibition of the proteasome did not
modulate the recruitment or cleavage of FADD, cFLIP, and
FIG. 9. MG115 sensitizes primary keratinocytes upstream or at the
mitochondrial level. (A) Cytochrome c, Smac/DIABLO, and HtrA2/
Omi release after TRAIL treatment of primary keratinocytes. Cells
were treated for the indicated time intervals with 50 ng of TRAIL per
ml in the presence or absence of 10 M MG115. Cytoplasmic lysates
were subsequently prepared as described in Materials and Methods
and analyzed for cytochrome c, Smac/DIABLO, and HtrA2/Omi by
Western blot analysis. Equal loading of cytoplasmic proteins was de-
termined by reprobing of blots with antitubulin antibodies.
(B) TRAIL-induced loss of mitochondrial transmembrane potential
⌬⌿
m
requires the presence of the proteasome inhibitor MG115. Cells
were incubated for 60 min with either diluent alone or 10 M MG115,
followed by treatment with 50 ng of TRAIL per ml. Cells were ana-
lyzed for ⌬⌿
m
by staining with 40 nM tetramethylrhodamine ethyl
ester 4 h later. Quantitative disruption of ⌬⌿
m
was only seen in cells
treated with TRAIL in the presence of the proteasome inhibitor
MG115.
786 LEVERKUS ET AL. M
OL.CELL.BIOL.
caspase 8 or 10 at the DISC, suggesting that the proteasome
inhibitor MG115 sensitizes primary keratinocytes downstream
of the TRAIL DISC. Intriguingly, active initiator caspases can
be detected in TRAIL-treated primary keratinocytes without
signicant apoptosis. Cleavage of caspase 3 to the 20-kDa
fragment is found at similar levels in cells treated with TRAIL
in the absence versus the presence of proteasome inhibitors.
These data further support the notion that caspase 8 is suf-
ciently activated to achieve initial cleavage of caspase 3. Thus,
our data indicate that besides the activity of the TRAIL DISC,
additional intracellular regulatory mechanisms exist. These
mechanisms control TRAIL-induced effector caspase activity
before a point of no return is passed, at which apoptosis
induction is inevitable. Importantly, in primary cells, as exem-
plied here for primary keratinocytes, low levels of initiator
caspase activation might be tolerated without induction of apo-
ptosis, thereby activating nonapoptotic signaling pathways.
To date, little is known about the signaling events associated
with TRAIL receptor ligation downstream of the DISC. In
particular, the role of mitochondrial signaling pathways re-
mains to be elucidated for TRAIL-induced apoptosis. In some
cell lines examined, TRAIL-induced apoptosis was shown to
be largely independent of mitochondrial signaling pathways
(32, 77). In other studies, Bcl-x
L
protected pancreatic carci
-
noma cells from TRAIL-induced apoptosis (24), and inactiva-
tion of Bax was recently shown to confer resistance to TRAIL-
induced apoptosis in colon cancer lines (41). Taken together,
these data support the view that cell type-specic differences
downstream of active initiator caspases might be important for
the cell fate decision following stimulation of TRAIL recep-
tors.
Our studies show that proteasome inhibitors induce major
and early differences in the TRAIL-dependent release of cy-
tochrome c, Smac/DIABLO, and HtrA2 (64) despite similar
early activation of initiator caspases. Importantly, the protea-
some inhibitor alone had no effect on mitochondrial cyto-
chrome c, Smac/DIABLO, or HtrA2 release which is in con-
trast to ndings obtained in transformed cell lines (75). This
difference might be due to a variable sensitivity of tumor cells
to proteasome inhibitors compared to primary cells (50). Our
data suggest that in primary keratinocytes, activation of the
mitochondrial apoptotic pathway is required for the execution
of TRAIL-induced apoptosis and that proteasome inhibitors
enable the release of proapoptotic molecules such as Smac/
DIABLO, cytochrome c, and HtrA2/Omi from mitochondria
in response to TRAIL treatment.
Our data showing partial cleavage of caspase 3 in primary
keratinocytes are consistent with recent reports demonstrating
that TRAIL-induced activation of Smac/DIABLO release is
dependent on Bax and Bak, promoted by Bid, and inhibited by
Bcl-2 and Bcl-x
L
downstream of caspase 8 activation in differ
-
ent tumor cell lines (12, 69). In line with these reports, the
release of Smac/DIABLO, HtrA2/Omi, and cytochrome c in
the presence of the proteasome inhibitor correlated with full
maturation of caspase 3 to the 17-kDa fragment in primary
keratinocytes. Interestingly, analysis of several Bcl-2 family
members, including Bcl-X
L
, Bax, and Bid, did not reveal
changes in the expression levels or cleavage patterns of these
molecules at time points when full caspase 3 maturation and
release of mitochondrial proapoptotic proteins is already de-
tected in primary keratinocytes (data not shown). Thus, it
remains to be determined if modulation of the expression
levels of Bcl-2 family members and their translocation to mi-
tochondria or other potential targets are modulated by protea-
some inhibitors.
Moreover, it is possible that cytochrome c-mediated activa-
tion of the apoptosome (5, 7) might also be involved in in-
creased caspase 3 activation. However, our data clearly show
that synergistic action of mitochondrion-derived proapoptotic
molecules in concert with death receptor-mediated caspase 3
activation is ultimately necessary and sufcient to overcome
inhibitory molecules such as XIAP in primary keratinocytes. In
support of this concept, recent in vitro experiments demon-
strated that an increase in Smac/DIABLO levels can overcome
XIAP-mediated inhibition of caspase 3 maturation (69). Of
interest, the removal of the prodomain of the effector caspase
6 is required for full enzymatic activity, further supporting our
ndings for caspase 3 (10).
When comparing the dose-dependent cleavage patterns of
TRAIL-resistant primary keratinocytes and highly sensitive
transformed keratinocytes, low concentrations of TRAIL were
sufcient to induce fully active p17 fragments in transformed
cells but not in primary cells, indicating that primary keratin-
ocytes require an additional signal for full caspase 3 matura-
tion. As shown in this study, one explanation for this difference
is the differential expression of XIAP, which inhibits the auto-
proteolytic maturation of caspase 3 (14, 15). Primary keratin-
ocytes might require the release of Smac/DIABLO and HtrA2/
Omi (64) from the mitochondria for inactivation of XIAP.
Thus, the stoichiometry of DISC-generated active caspase 8,
cytoplasmic XIAP, and mitochondrion-released Smac/DIA-
BLO or HtrA2/Omi might dene resistance or sensitivity to
TRAIL-induced apoptosis in primary keratinocytes and possi-
bly also in other cells, as suggested recently (69).
Remarkably, there may be important cell type-specic dif-
ferences in the action of proteasome inhibitors, as primary
human hepatocytes are not sensitized to TRAIL by these
agents (T. Ganten and H. Walczak, unpublished data). These
differences might be explained by differential expression of
TRAIL receptors, distinct DISC components, or downstream
inhibitory molecules, as described in this report. Our data
demonstrating interaction of XIAP with caspase 3 under phys-
iological conditions as well as increased resistance to TRAIL-
induced apoptosis following ectopic expression of XIAP in
highly TRAIL-sensitive transformed keratinocytes further sup-
port the important role of XIAP in TRAIL resistance in pri-
mary keratinocytes.
Taken together, our data support a model in which pro-
teasome inhibitors allow initiator caspase-mediated activa-
tion of mitochondrial pathways of apoptosis in primary cells.
The activation of mitochondrial pathways is necessary for
inactivation of XIAP and, consequently, of full caspase 3
activity in primary keratinocytes (Fig. 10). Alternatively,
XIAP might exert its caspase-inhibitory role by its ubiquitin-
protein ligase activity (70). Since polyubiquitination labels
proteins for proteasomal degradation, proteasome inhibi-
tion may thereby lead to increased levels of active caspase 3
and subsequent apoptotic cell death in cells expressing high
levels of XIAP. This mechanism might not be required in
cells expressing low levels of XIAP, as supported by our
VOL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 787
overexpression studies. However, another possibility of the
action of the proteasomal inhibitor may be decreased XIAP-
mediated degradation of Smac/DIABLO, as suggested by a
very recent report (47). Future studies are required to de-
lineate these points.
It is possible that the resistance mechanism of primary
human keratinocytes outlined in this study is also relevant
for other types of primary cells and could thus be a more
general mechanism of resistance of primary cells to TRAIL.
In that respect, our ndings suggest that the use of protea-
some inhibitors as anticancer agents might lead to unwanted
sensitization of primary cells to death receptor-mediated
apoptosis. Thus, the clinical application of proteasome in-
hibitors, either alone or in combination with death receptor
agonists such as TRAIL, must be closely monitored for
unwanted toxicity that could be due to the apoptosis-sensi-
tizing effect exerted by these agents on primary cells as
identied in this study.
ACKNOWLEDGMENTS
We thank P. H. Krammer for monoclonal antibodies to caspase 8
(C-15) and cFLIP (NF-6), D. Nicholson for caspase 3 and XIAP
antisera as well as biotin-DEVD-aomk, M. MacFarlane for rabbit
serum to Smac/DIABLO, X. Wang for rabbit serum to Bid, R. Taka-
hashi for rabbit serum to HtrA2/Omi, S. Roy for the protocol to
perform afnity labeling experiments, and C. S. Duckett for the XIAP
expression vector. We are also grateful to Evi Horn for excellent
technical assistance and Heiko Stahl for generating monoclonal anti-
bodies to TRAIL-R1 to -R4.
Part of this study was funded by grants from the IZKF Wu¨rzburg
(01 K5 9603), Sander-Stiftung (2000.092.1), and Deutsche Kreb-
shilfe (10-1951-Le1) to Martin Leverkus. Henning Walczak is sup-
ported by a BioFuture grant from the Bundesministerium fu¨r Bil-
dung und Forschung (BMBF). Manfred Neumann and Bernd
Baumann were supported by grants from the Deutsche Forschungs-
gemeinschaft (NE 608/1-3 and/2-2 to M.N. and SFB 497/B1 to
B.B.).
The rst two authors contributed equally to this paper.
The last two authors share senior authorship of this paper.
FIG. 10. Model of action of proteasome inhibitors mediating sensitization of primary keratinocytes to TRAIL. TRAIL activates caspases 8 and
10 at the DISC. Initial processing of caspase 3 produces a p20/p12 intermediate of caspase 3 which is inhibited by XIAP. Proteasome inhibitors
interfere with caspase 8-mediated activation of mitochondrial pathways of apoptosis, leading to the caspase 8-activated release of Smac/DIABLO
and HtrA2/Omi and subsequent removal of the caspase 3 prodomain (hatched bars). This activation of mitochondrial pathways is necessary for
inactivation of XIAP and subsequent full caspase 3 activity in primary keratinocytes.
788 LEVERKUS ET AL. M
OL.CELL.BIOL.
REFERENCES
1. Ashkenazi, A., R. C. Pai, S. Fong, S. Leung, D. A. Lawrence, S. A. Marsters,
C. Blackie, L. Chang, A. E. McMurtrey, A. Hebert, L. DeForge, I. L. Kou-
menis, D. Lewis, L. Harris, J. Bussiere, H. Koeppen, Z. Shahrokh, and R. H.
Schwall. 1999. Safety and antitumor activity of recombinant soluble Apo2
ligand. J. Clin. Investig. 104:155162.
2. Beg, A. A., W. C. Sha, R. T. Bronson, S. Ghosh, and D. Baltimore. 1995.
Embryonic lethality and liver degeneration in mice lacking the RelA com-
ponent of NF-B. Nature 376:167170.
3. Bodmer, J. L., N. Holler, S. Reynard, P. Vinciguerra, P. Schneider, P. Juo,
J. Blenis, and J. Tschopp. 2000. TRAIL receptor-2 signals apoptosis through
FADD and caspase-8. Nat. Cell Biol. 2:241243.
4. Boukamp, P., R. T. Petrussevska, D. Breitkreutz, J. Hornung, A. Markham,
and N. E. Fusenig. 1988. Normal keratinization in a spontaneously immor-
talized aneuploid human keratinocyte cell line. J. Cell Biol. 106:761771.
5. Bratton, S. B., G. Walker, S. M. Srinivasula, X. M. Sun, M. Butterworth,
E. S. Alnemri, and G. M. Cohen. 2001. Recruitment, activation and retention
of caspases-9 and -3 by Apaf-1 apoptosome and associated XIAP complexes.
EMBO J. 20:9981009.
6. Briegel, K., B. Hentsch, I. Pfeuffer, and E. Sering. 1991. One base pair
change abolishes the T cell-restricted activity of a B-like proto-enhancer
element from the interleukin 2 promoter. Nucleic Acids Res. 19:59295936.
7. Cain, K., D. G. Brown, C. Langlais, and G. M. Cohen. 1999. Caspase
activation involves the formation of the aposome, a large (approximately 700
kDa) caspase-activating complex. J. Biol. Chem. 274:2268622692.
8. Chai, J., C. Du, J. W. Wu, S. Kyin, X. Wang, and Y. Shi. 2000. Structural and
biochemical basis of apoptotic activation by Smac/DIABLO. Nature 406:
855862.
9. Chen, F., D. Chang, M. Goh, S. A. Klibanov, and M. Ljungman. 2000. Role
of p53 in cell cycle regulation and apoptosis following exposure to protea-
some inhibitors. Cell Growth Differ. 11:239246.
10. Cowling, V., and J. Downward. 2002. Caspase 6 is the direct activator of
caspase 8 in the cytochrome c-induced apoptosis pathway: absolute require-
ment for removal of caspase 6 prodomain. Cell Death Differ. 9:10461056.
11. Degli-Esposti, M. A., W. C. Dougall, P. J. Smolak, J. Y. Waugh, C. A. Smith,
and R. G. Goodwin. 1997. The novel receptor TRAIL-R4 induces NF-B and
protects against TRAIL-mediated apoptosis, yet retains an incomplete death
domain. Immunity 7:813820.
12. Deng, Y., Y. Lin, and X. Wu. 2002. TRAIL-induced apoptosis requires
Bax-dependent mitochondrial release of Smac/DIABLO. Genes Dev. 16:33
45.
13. Denk, A., M. Goebeler, S. Schmid, I. Berberich, O. Ritz, D. Lindemann, S.
Ludwig, and T. Wirth. 2001. Activation of NF-B via the IB kinase complex
is both essential and sufcient for proinammatory gene expression in pri-
mary endothelial cells. J. Biol. Chem. 276:2845128458.
14. Deveraux, Q. L., and J. C. Reed. 1999. IAP family proteinssuppressors of
apoptosis. Genes Dev. 13:239252.
15. Deveraux, Q. L., R. Takahashi, G. S. Salvesen, and J. C. Reed. 1997. X-
linked IAP is a direct inhibitor of cell-death proteases. Nature 388:300304.
16. Drexler, H. C. 1997. Activation of the cell death program by inhibition of
proteasome function. Proc. Natl. Acad. Sci. USA 94:855860.
17. Du, C., M. Fang, Y. Li, L. Li, and X. Wang. 2000. Smac, a mitochondrial
protein that promotes cytochrome c-dependent caspase activation by elimi-
nating IAP inhibition. Cell 102:3342.
18. Ekert, P. G., J. Silke, C. J. Hawkins, A. M. Verhagen, and D. L. Vaux. 2001.
DIABLO promotes apoptosis by removing MIHA/XIAP from processed
caspase 9. J. Cell Biol. 152:483490.
19. Fenteany, G., R. F. Standaert, W. S. Lane, S. Choi, E. J. Corey, and S. L.
Schreiber. 1995. Inhibition of proteasome activities and subunit-specic ami-
no-terminal threonine modication by lactacystin. Science 268:726731.
20. Gliniak, B., and T. Le. 1999. Tumor necrosis factor-related apoptosis-induc-
ing ligands antitumor activity in vivo is enhanced by the chemotherapeutic
agent CPT-11. Cancer Res. 59:61536158.
21. Goldstein, J. C., N. J. Waterhouse, P. Juin, G. I. Evan, and D. R. Green.
2000. The coordinate release of cytochrome c during apoptosis is rapid,
complete and kinetically invariant. Nat. Cell Biol. 2:156162.
22. Grifth, T. S., W. A. Chin, G. C. Jackson, D. H. Lynch, and M. Z. Kubin.
1998. Intracellular regulation of TRAIL-induced apoptosis in human mela-
noma cells. J. Immunol. 161:28332840.
23. Harper, N., S. N. Farrow, A. Kaptein, G. M. Cohen, and M. MacFarlane.
2001. Modulation of TRAIL-induced NF-B activation by inhibition of api-
cal caspases. J. Biol. Chem. 276:3474334752.
24. Hinz, S., A. Trauzold, L. Boenicke, C. Sandberg, S. Beckmann, E. Bayer, H.
Walczak, H. Kalthoff, and H. Ungefroren. 2000. Bcl-XL protects pancreatic
adenocarcinoma cells against CD95- and TRAIL-receptor-mediated apopto-
sis. Oncogene 19:54775486.
25. Holcik, M., and R. G. Korneluk. 2001. XIAP, the guardian angel. Nat. Rev.
Mol. Cell. Biol. 2:550556.
26. Hu, W. H., H. Johnson, and H. B. Shu. 1999. Tumor necrosis factor-related
apoptosis-inducing ligand receptors signal NF-B and JNK activation and
apoptosis through distinct pathways. J. Biol. Chem. 274:3060330610.
27. Jeremias, I., C. Kupatt, B. Baumann, I. Herr, T. Wirth, and K. M. Debatin.
1998. Inhibition of nuclear factor B activation attenuates apoptosis resis-
tance in lymphoid cells. Blood 91:46244631.
28. Jesenberger, V., and S. Jentsch. 2002. Deadly encounter: ubiquitin meets
apoptosis. Nat. Rev. Mol. Cell. Biol. 3:112121.
29. Karin, M., and Y. Ben-Neriah. 2000. Phosphorylation meets ubiquitination:
the control of NF-B activity. Ann. Rev. Immunol. 18:621663.
30. Karin, M., and A. Lin. 2002. NF-B at the crossroads of life and death. Nat.
Immunol. 3:221227.
31. Kataoka, T., R. C. Budd, N. Holler, M. Thome, F. Martinon, M. Irmler, K.
Burns, M. Hahne, N. Kennedy, M. Kovacsovics, and J. Tschopp. 2000. The
caspase-8 inhibitor FLIP promotes activation of NF-B and Erk signaling
pathways. Curr. Biol. 10:640648.
32. Keogh, S. A., H. Walczak, L. Bouchier-Hayes, and S. J. Martin. 2000. Failure
of Bcl-2 to block cytochrome c redistribution during TRAIL-induced apo-
ptosis. FEBS Lett. 471:9398.
33. Kischkel, F. C., S. Hellbardt, I. Behrmann, M. Germer, M. Pawlita, P. H.
Krammer, and M. E. Peter. 1995. Cytotoxicity-dependent APO-1 (Fas/
CD95)-associated proteins form a death-inducing signaling complex (DISC)
with the receptor. EMBO J. 14:55795588.
34. Kischkel, F. C., D. A. Lawrence, A. Chuntharapai, P. Schow, K. J. Kim, and
A. Ashkenazi. 2000. Apo2L/TRAIL-dependent recruitment of endogenous
FADD and caspase-8 to death receptors 4 and 5. Immunity 12:611620.
35. Kischkel, F. C., D. A. Lawrence, A. Tinel, H. LeBlanc, A. Virmani, P. Schow,
A. Gazdar, J. Blenis, D. Arnott, and A. Ashkenazi. 2001. Death receptor
recruitment of endogenous caspase-10 and apoptosis initiation in the ab-
sence of caspase-8. J. Biol. Chem. 276:4663946646.
36. Koepp, D. M., J. W. Harper, and S. J. Elledge. 1999. How the cyclin became
a cyclin: regulated proteolysis in the cell cycle. Cell 97:431434.
37. Kroemer, G., and J. C. Reed. 2000. Mitochondrial control of cell death. Nat.
Med. 6:513951.
38. Krueger, A., I. Schmitz, S. Baumann, P. H. Krammer, and S. Kirchhoff.
2001. Cellular FLICE-inhibitory protein splice variants inhibit different steps
of caspase-8 activation at the CD95 death-inducing signaling complex.
J. Biol. Chem. 276:2063320640.
39. Laney, J. D., and M. Hochstrasser. 1999. Substrate targeting in the ubiquitin
system. Cell 97:427430.
40. Lazebnik, Y. A., S. H. Kaufmann, S. Desnoyers, G. G. Poirier, and W. C.
Earnshaw. 1994. Cleavage of poly(ADP-ribose) polymerase by a proteinase
with properties like ICE. Nature 371:346347.
41. LeBlanc, H., D. A. Lawrence, E. Varfolomeev, K. Totpal, J. Morlan, P.
Schow, S. Fong, R. Schwall, D. Sinicropi, and A. Ashkenazi. 2002. Tumor-
cell resistance to death receptor-induced apoptosis through mutational in-
activation of the proapoptotic Bcl-2 homolog Bax. Nat. Med. 8:274281.
42. Leverkus, M., M. Neumann, T. Mengling, C. T. Rauch, E. B. Bro¨cker, P. H.
Krammer, and H. Walczak. 2000. Regulation of TRAIL sensitivity in pri-
mary and transformed human keratinocytes. Cancer Res. 60:553559.
43. Leverkus, M., H. Walczak, H.-W. Fries, G. Terbeck, A. McLellan, E.-B.
Bro¨cker, and E. Ka¨mpgen. 2000. Maturation of dendritic cells leads to
upregulation of cellular FLICE-inhibitory protein (cFLIP) and concomitant
downregulation of death receptor mediated apoptosis. Blood 110:353357.
44. Leverkus, M., M. Yaar, and B. A. Gilchrest. 1997. Fas/Fas ligand interaction
contributes to UV-induced apoptosis in human keratinocytes. Exp. Cell Res.
232:255262.
45. Liu, Z., C. Sun, E. T. Olejniczak, R. P. Meadows, S. F. Betz, T. Oost,
J. Herrmann, J. C. Wu, and S. W. Fesik. 2000. Structural basis for binding of
Smac/DIABLO to the XIAP BIR3 domain. Nature 408:10041008.
46. Locksley, R. M., N. Killeen, and M. J. Lenardo. 2001. The tumor necrosis
factor and tumor necrosis factor receptor superfamilies: integrating mam-
malian biology. Cell 104:487501.
47. MacFarlane, M., W. Merrison, S. B. Bratton, and G. M. Cohen. 2002.
Proteasome-mediated degradation of Smac during apoptosis:XIAP pro-
motes Smac ubiquitination in vitro. J. Biol. Chem. 277:3661136616.
48. Marienfeld, R., F. Berberich-Siebelt, I. Berberich, A. Denk, E. Sering, and
M. Neumann. 2001. Signal-specic and phosphorylation-dependent RelB
degradation: a potential mechanism of NF-B control. Oncogene 20:8142
8147.
49. Martins, L. M. 2002. The serine protease Omi/HtrA2: a second mammalian
protein with a Reaper-like function. Cell Death Differ. 9:699701.
50. Masdehors, P., H. Merle-Beral, H. Magdelenat, and J. Delic. 2000. Ubiq-
uitin-proteasome system and increased sensitivity of B-CLL lymphocytes to
apoptotic death activation. Leukemia Lymphoma 38:499504.
51. Medema, J. P., C. Scafdi, P. H. Krammer, and M. E. Peter. 1998. Bcl-xL
acts downstream of caspase-8 activation by the CD95 death-inducing signal-
ing complex. J. Biol. Chem. 273:33883393.
52. Meng, L., R. Mohan, B. H. Kwok, M. Elofsson, N. Sin, and C. M. Crews.
1999. Epoxomicin, a potent and selective proteasome inhibitor, exhibits in
vivo antiinammatory activity. Proc. Natl. Acad. Sci. USA 96:1040310408.
53. Neumann, M., T. Grieshammer, S. Chuvpilo, B. Kneitz, M. Lohoff, A.
Schimpl, B. R. Franza, Jr., and E. Sering. 1995. RelA/p65 is a molecular
target for the immunosuppressive action of protein kinase A. EMBO J.
14:19912004.
VOL. 23, 2003 EFFECTOR CASPASE INHIBITION IN PRIMARY KERATINOCYTES 789
54. Nicholson, D. W. 1999. Caspase structure, proteolytic substrates, and func-
tion during apoptotic cell death. Cell Death Differ. 6:10281042.
55. Palombella, V. J., O. J. Rando, A. L. Goldberg, and T. Maniatis. 1994. The
ubiquitin-proteasome pathway is required for processing the NF-B1 pre-
cursor protein and the activation of NF-B. Cell 78:773785.
56. Pitti, R. M., S. A. Marsters, S. Ruppert, C. J. Donahue, A. Moore, and A.
Ashkenazi. 1996. Induction of apoptosis by Apo-2 ligand, a new member of
the tumor necrosis factor cytokine family. J. Biol. Chem. 271:1268712690.
57. Roy, S., C. I. Bayly, Y. Gareau, V. M. Houtzager, S. Kargman, S. L. C. Keen,
K. Rowland, I. M. Seiden, N. A. Thornberry, and D. W. Nicholson. 2001.
Maintenance of caspase-3 proenzyme dormancy by an intrinsic safety
catch regulatory tripeptide. Proc. Natl. Acad. Sci. USA 98:61326137.
58. Roy, S., and D. W. Nicholson. 2000. Cross-talk in cell death signalling. J. Exp.
Med. 192:F21F25.
59. Scaduto, R. C., Jr., and L. W. Grotyohann. 1999. Measurement of mitochon-
drial membrane potential with uorescent rhodamine derivatives. Biophys. J.
76:469477.
60. Scafdi, C., S. Fulda, A. Srinivasan, C. Friesen, F. Li, K. J. Tomaselli, K. M.
Debatin, P. H. Krammer, and M. E. Peter. 1998. Two CD95 (APO-1/Fas)
signaling pathways. EMBO J. 17:16751687.
61. Scafdi, C., I. Schmitz, P. H. Krammer, and M. E. Peter. 1999. The role of
c-FLIP in modulation of CD95-induced apoptosis. J. Biol. Chem. 274:1541
1548.
62. Schneider, P., M. Thome, K. Burns, J. L. Bodmer, K. Hofmann, T. Kataoka,
N. Holler, and J. Tschopp. 1997. TRAIL receptors 1 (DR4) and 2 (DR5)
signal FADD-dependent apoptosis and activate NF-B. Immunity 7:831
836.
63. Schreiber, E., P. Matthias, M. M. Muller, and W. Schaffner. 1989. Rapid
detection of octamer binding proteins with mini-extracts, prepared from a
small number of cells. Nucleic Acids Res. 17:64196424.
64. Shi, Y. 2002. Mechanisms of caspase activation and inhibition during apo-
ptosis. Mol. Cell 9:459470.
65. Silke, J., P. G. Ekert, C. L. Day, C. J. Hawkins, M. Baca, J. Chew, M.
Pakusch, A. M. Verhagen, and D. L. Vaux. 2001. Direct inhibition of caspase
3 is dispensable for the anti-apoptotic activity of XIAP. EMBO J. 20:3114
3123.
66. Single, B., M. Leist, and P. Nicotera. 1998. Simultaneous release of adenyl-
ate kinase and cytochrome c in cell death. Cell Death Differ. 5:10011003.
67. Sprick, M. R., E. Rieser, H. Stahl, A. Grosse-Wilde, M. A. Weigand, and H.
Walczak. 2002. Caspase 10 is recruited to and activated at the native TRAIL
and CD95 death-inducing signalling complexes in a FADD-dependent man-
ner. EMBO J. 21:45204530.
68. Sprick, M. R., M. A. Weigand, E. Rieser, C. T. Rauch, P. Juo, J. Blenis, P. H.
Krammer, and H. Walczak. 2000. FADD/MORT1 and caspase-8 are re-
cruited to TRAIL receptors 1 and 2 and are essential for apoptosis mediated
by TRAIL receptor 2. Immunity 12:599609.
69. Sun, X. M., S. B. Bratton, M. Butterworth, M. MacFarlane, and G. M.
Cohen. 2002. Bcl-2 and Bcl-xL inhibit CD95-mediated apoptosis by prevent-
ing mitochondrial release of Smac/DIABLO and subsequent inactivation of
X-linked inhibitor-of-apoptosis protein. J. Biol. Chem. 277:1134511351.
70. Suzuki, Y., Y. Nakabayashi, and R. Takahashi. 2001. Ubiquitin-protein
ligase activity of X-linked inhibitor of apoptosis protein promotes proteaso-
mal degradation of caspase-3. Proc. Natl. Acad. Sci. USA 98:86628667.
71. Thompson, C. B. 1995. Apoptosis in the pathogenesis and treatment of
disease. Science 267:14561562.
72. Thornberry, N. A., E. P. Peterson, J. J. Zhao, A. D. Howard, P. R. Grifn,
and K. T. Chapman. 1994. Inactivation of interleukin-1 beta converting
enzyme by peptide (acyloxy)methyl ketones. Biochemistry 33:39343940.
73. van Loo, G., X. Saelens, M. van Gurp, M. MacFarlane, S. J. Martin, and P.
Vandenabeele. 2002. The role of mitochondrial factors in apoptosis: a Rus-
sian roulette with more than one bullet. Cell Death Differ. 9:10311042.
74. Verhagen, A. M., P. G. Ekert, M. Pakusch, J. Silke, L. M. Connolly, G. E.
Reid, R. L. Moritz, R. J. Simpson, and D. L. Vaux. 2000. Identication of
DIABLO, a mammalian protein that promotes apoptosis by binding to and
antagonizing IAP proteins. Cell 102:4353.
75. Wagenknecht, B., M. Hermisson, P. Groscurth, P. Liston, P. H. Krammer,
and M. Weller. 2000. Proteasome inhibitor-induced apoptosis of glioma cells
involves the processing of multiple caspases and cytochrome c release.
J. Neurochem. 75:22882297.
76. Wajant, H., E. Haas, R. Schwenzer, F. Muhlenbeck, S. Kreuz, G. Schubert,
M. Grell, C. Smith, and P. Scheurich. 2000. Inhibition of death receptor-
mediated gene induction by a cycloheximide-sensitive factor occurs at the
level of or upstream of Fas-associated death domain protein (FADD).
J. Biol. Chem. 275:2435724366.
77. Walczak, H., A. Bouchon, H. Stahl, and P. H. Krammer. 2000. Tumor
necrosis factor-related apoptosis-inducing ligand retains its apoptosis-induc-
ing capacity on Bcl-2- or Bcl-xL-overexpressing chemotherapy-resistant tu-
mor cells. Cancer Res. 60:30513057.
78. Walczak, H., M. A. Degli-Esposti, R. S. Johnson, P. J. Smolak, J. Y. Waugh,
N. Boiani, M. S. Timour, M. J. Gerhart, K. A. Schooley, C. A. Smith, R. G.
Goodwin, and C. T. Rauch. 1997. TRAIL-R2: a novel apoptosis-mediating
receptor for TRAIL. EMBO J. 16:53865397.
79. Walczak, H., and P. H. Krammer. 2000. The CD95 (APO-1/Fas) and the
TRAIL (APO-2L) apoptosis systems. Exp. Cell Res. 256:5866.
80. Walczak, H., R. E. Miller, K. Ariail, B. Gliniak, T. S. Grifth, M. Kubin, W.
Chin, J. Jones, A. Woodward, T. Le, C. Smith, P. Smolak, R. G. Goodwin,
C. T. Rauch, J. C. Schuh, and D. H. Lynch. 1999. Tumoricidal activity of
tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat. Med.
5:157163.
81. Walczak, H., and M. R. Sprick. 2001. Biochemistry and function of the
DISC. Trends Biochem. Sci. 26:452453.
82. Wallach, D., E. E. Varfolomeev, N. L. Malinin, Y. V. Goltsev, A. V.
Kovalenko, and M. P. Boldin. 1999. Tumor necrosis factor receptor and Fas
signaling mechanisms. Annu. Rev. Immunol. 17:331367.
83. Wang, C. Y., M. W. Mayo, R. G. Korneluk, D. V. Goeddel, and A. S. Baldwin,
Jr. 1998. NF-B antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1
and c-IAP2 to suppress caspase-8 activation. Science 281:16801683.
84. Wang, J., H. J. Chun, W. Wong, D. M. Spencer, and M. J. Lenardo. 2001.
Caspase-10 is an initiator caspase in death receptor signaling. Proc. Natl.
Acad. Sci. USA 98:1388413888.
85. Wehrli, P., I. Viard, R. Bullani, J. Tschopp, and L. E. French. 2000. Death
receptors in cutaneous biology and disease. J. Investig. Dermatol. 115:141
148.
86. Wiley, S. R., K. Schooley, P. J. Smolak, W. S. Din, C. P. Huang, J. K. Nicholl,
G. R. Sutherland, T. D. Smith, C. Rauch, C. A. Smith, and R. G. Goodwin.
1995. Identication and characterization of a new member of the tumor
necrosis factor family that induces apoptosis. Immunity 3:673682.
87. Wu, M. X., Z. Ao, K. V. Prasad, R. Wu, and S. F. Schlossman. 1998. IEX-1L,
an apoptosis inhibitor involved in NF-B-mediated cell survival. Science
281:9981001.
88. Zhao, J., T. Tenev, L. M. Martins, J. Downward, and N. R. Lemoine. 2000.
The ubiquitin-proteasome pathway regulates survivin degradation in a cell
cycle-dependent manner. J. Cell Sci. 113:43634371.
790 LEVERKUS ET AL. MOL.CELL.BIOL.
... , Budd et al. 2006. In bisherigen Arbeiten erfolgte üblicherweise der Nachweis von cFLIP mittels Western Blot Analysen (Irmler et al. 1997, Griffith et al. 1998, Scaffidi et al. 1999, Kamarajan et al. 2003, Leverkus et al. 2003 (Scaffidi et al. 1999, Leverkus et al. 2003, Wachter et al. 2004, Kavuri et al. 2011 ...
... , Budd et al. 2006. In bisherigen Arbeiten erfolgte üblicherweise der Nachweis von cFLIP mittels Western Blot Analysen (Irmler et al. 1997, Griffith et al. 1998, Scaffidi et al. 1999, Kamarajan et al. 2003, Leverkus et al. 2003 (Scaffidi et al. 1999, Leverkus et al. 2003, Wachter et al. 2004, Kavuri et al. 2011 ...
Thesis
Die Resistenz von Tumorzellen gegenüber Apoptose stellt einen zentralen Baustein in der Pathogenese von Tumorerkrankungen dar. cFLIP inhibiert rezeptornah die Todesrezeptor-vermittelte Apoptose und spielt somit eine bedeutende Rolle als Regulator der Apoptose. Eine verstärkte Expression von cFLIP kann folglich hinweisend auf eine Fehlregulation der Apoptose bei der Entstehung und Progression von Tumoren sein. In der vorliegenden Arbeit wurde die Expression von cFLIP in kutanen epithelialen und melanozytären Tumoren mit formalinfixierten und paraffinierten Gewebeproben untersucht. Bei der zunächst durchgeführten Charakterisierung der käuflich erhältlichen monoklonalen cFLIP-Antikörper mittels cFLIP-überexprimierenden HaCaT-Keratinozyten wurde überraschenderweise die fehlende Spezifität eines Antikörpers (KlonEPR8438(2)) nachgewiesen, was zur Folge hatte, dass der Hersteller die Produktion nach Mitteilung der Befunde eingestellt hat. Daher wurden die weiteren Untersuchungen unter Verwendung des Antikörper-Klons G11, der sowohl im Western Blot als auch immunhistochemisch die beiden cFLIP-Splicevarianten cFLIPL und cFLIPS spezifisch nachweist, durchgeführt. Hierbei konnte gezeigt werden, dass cFLIP in epithelialen Hauttumoren in erheblichem Maß exprimiert wird. In jeweils 40% der untersuchten aktinischen Keratosen und Morbi Bowen konnte cFLIP nachgewiesen werden. Im Vergleich dazu zeigte sich in den fortgeschrittenen Formen epithelialer Hauttumoren eine deutlich höhere Expressionsrate. Eine Expression wiesen zudem 100% der untersuchten Keratoakanthome und 95% der Plattenepithelkarzinome (mit überwiegend gutem Differenzierungsgrad) auf. Dementsprechend war es nicht verwunderlich, dass alle untersuchten Metastasen von Plattenepithelkarzinomen cFLIP überexprimierten. Die Analyse melanozytärer Läsionen ergab, dass cFLIP in melanozytären Nävi wie auch superfiziell spreitenden Melanomen, Lentigo-maligna-Melanomen und akral lentiginösen Melanomen nur in einer sehr geringen Anzahl der untersuchten Präparate überexprimiert wurde. Erstaunlicherweise konnte jedoch in 65% der nodulären Melanome sowie in 60% der Melanommetastasen cFLIP nachgewiesen werden. Bezüglich der Expression von cFLIP im Primärtumor sowie der Metastase desselben Patienten konnte kein eindeutiger Trend festgestellt werden. Die Ergebnisse dieser Arbeit deuten darauf hin, dass die frühe Hemmung des extrinsischen Apoptoseweges durch das antiapoptotische Protein cFLIP an der Entstehung, dem Wachstum und möglicherweise der Metastasierung epithelialer Hauttumore beteiligt sein dürfte. Die auffallend hohe Expressionsrate im nodulären Melanom sowie den untersuchten Melanommetastasen könnte einen zukünftigen therapeutischen Angriffspunkt darstellen.
... To further understand the physiological role of cIAPs for CD95-induced cell death, we studied the treatment of cells with TWEAK, which was recently shown to induce cIAP in keratinocytes (Leverkus et al., 2003b), which is similar to the role of XIAP in type II cells (Jost et al., 2009). TWEAK-induced sensitization similarly uncovered a caspaseindependent, RIP1-dependent form of cell death (Fig. 8 C). ...
... HRP-conjugated goat antirabbit, goat anti-rat IgG, and goat anti-mouse IgG antibodies and HRPconjugated goat anti-mouse IgG1, IgG2a, IgG2b, and IgG1-were obtained from SouthernBiotech. TRAIL-R1 (HS 101) and TRAIL-R2 (HS 201) monoclonal antibodies for FACScan analysis of surface receptor expression were used as previously described (Leverkus et al., 2003b) and are available from Enzo Life Sciences, Inc. CD95 antibodies (Apo-1 IgG1 and IgG3a) were provided by P.H. Krammer. Cy5-conjugated annexin V was purchased from BD. ...
... La caspase-8 active ainsi que de nombreuses autres molécules pro-apoptotiques entrainent une meilleure sensibilité aux ligands de mort une fois qu'elles ne sont plus dégradées (de Wilt et al., 2013). Cependant, des études ont démontré que l'inhibition du protéasome en combinaison d'un traitement TRAIL aurait permis de sensibiliser des cellules saines primaires (kératinocytes) (Leverkus et al., 2003). ...
Thesis
L'hétérogénéité phénotypique observée dans des populations de cellules isogéniques est une cause immédiate de résistance non-génétique aux anticancéreux qui reste difficile à définir en raison de sa nature transitoire. Dans une première partie, nous avons étudié l’origine moléculaire de l'hétérogénéité phénotypique de réponse au TNF-related apoptosis-inducing ligand (TRAIL). Pour cela nous avons développé une méthode permettant d’isoler les cellules résistantes avant qu’elles n’engagent une induction génétique par le traitement, et les cellules sensibles avant qu’elles ne détériorent leur contenu lors de la mort cellulaire. La méthode emploie trois technologies en cellule unique : une mesure prédictive de réponse cellulaire par vidéo-microscopie, une isolation de chaque cellule in situ par capture laser, suivi d’un profilage transcriptomique. La prédiction de réponse thérapeutique permet alors d’isoler et profiler toutes les cellules, quelle que soit l’issue de leur réponse pharmacologique tardive (mort cellulaire pour les sensibles), et ainsi nous a permis d’identifier les gènes à l’origine de la différence d’efficacité de TRAIL dans une population de cellules clonales. Puis dans un second projet, nous avons étudié la résistance transitoire induite lors de traitement par TRAIL. Nous avons pu mettre en évidence le rôle de TAK-1 dans la mise en place de cette résistance transitoire. Il a été décrit que la voie apoptotique déclenchée par TRAIL pouvait aussi bifurquer sur la voie de la nécroptose, une mort cellulaire fortement immunogène. Nous avons pu démontrer une communication antagoniste de ces voies : la résistance induite lors de l’apoptose des cellules traitées par TRAIL permettait de sensibiliser les cellules survivantes au traitement nécroptotique. Ces travaux pourraient permettre de suggérer de nouvelles stratégies thérapeutiques afin de pallier les résistances inhérentes à ces voies de signalisations.
... Oncogenic Ras transformation of human fibroblasts has been shown to confer TRAIL resistance and this was associated with an enhanced recruitment of pro-caspase 8 to the DISC [164] and induction of the pro-apoptotic protein Bak [165]. Similar findings have been observed in transformed keratinocytes where increased TRAIL sensitivity was associated with both reduced cFLIP and XIAP expression [166,167]. A balance of pro-and anti-apoptotic factors expressed therefore ultimately determines whether a cell is TRAIL sensitive. ...
Conference Paper
Malignant pleural mesothelioma (MPM) is a devastating disease for which limited effective therapies are currently available. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and other death receptor (DR) agonists are pro-apoptotic agents that trigger the extrinsic apoptotic pathway selectively in cancer cells. Previous work has shown that supports that loss of function of the nuclear deubiquitinase BRCA associated protein-1 (BAP1) augments sensitivity to recombinant TRAIL (rTRAIL) in MPM cells. This study shows that BAP1 is a candidate biomarker for rTRAIL sensitivity in cell line, early passage culture and tumour explant models of MPM. In addition, BAP1 is a potential biomarker for sensitivity to other DR agonists and for additional cancer types with BAP1 mutations. I have also identified other novel candidate biomarkers for DR agonist sensitivity, notably ASXL1/2, through exploration of the mechanism underlying the BAP1-rTRAIL association. I present data supporting the clinical relevance and utility of the BAP1-TRAIL association. I have shown that loss of BAP1 function occurs in a significant proportion of MPM tumours in the UK and that loss of BAP1 function augments sensitivity to TRAIL in primary tumour tissue. I describe in vitro data supporting the hypothesis that loss of BAP1 function augments sensitivity to rTRAIL in MPM. I have shown that loss of BAP1 function augments sensitivity to other DR agonists and that BAP1 can act as a biomarker for DR agonist sensitivity in other cancers with BAP1 mutations. Finally, I explore the mechanism underlying the BAP1-TRAIL association. I present data supporting the notion that BAP1 binds to the ASXL1/2 proteins to form the polycomb repressor deubiquitinase complex which underlies BAP1-induced TRAIL resistance. Loss of this function results in a change in the expression of proteins of the extrinsic apoptotic pathway, which may favour apoptosis upon DR agonist binding.
... [29] The two pathways converge at caspase-3 activation; thus, caspase-3 is considered a key enzyme in the pathogenesis of cell apoptosis. [30][31][32] The level of cleaved caspase-3 represents the level of activated caspase-3. [33,34] High levels of cleaved caspase-3 in certain tumor may predict good survival. ...
Article
Full-text available
Background: The suillin isoform iso-suillin is a natural substance isolated from a petroleum ether extract of the fruiting bodies of the mushroom Suillus flavus. Previous studies have found its inhibition effect on some cancer cells, and we aimed to study its effects on human small cell lung cancer H446 cell line. Methods: Cell viability was measured by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Cellular morphological changes (apoptosis and necrosis) were evaluated using an electron microscope and Hoechst 33258 staining detected by the inverted microscope. Flow cytometry was used to detect cell apoptosis, cell cycle distribution, and mitochondrial membrane potential. Protein expression was determined by Western blotting analysis. Results: Here, we describe the ability of iso-suillin to inhibit the growth of H446 cells in time- and dose-dependent way. Iso-suillin had no obvious impact on normal human lymphocyte proliferation at low concentrations (9.09, 18.17, or 36.35 μmol/L) but promoted lymphocyte proliferation at a high concentration (72.70 μmol/L). After treatment of different concentrations of iso-suillin (6.82, 13.63, or 20.45 μmol/L), the apoptosis rate of H446 cells increased with increasing concentrations of iso-suillin (16.70%, 35.54%, and 49.20%, respectively, all P < 0.05 compared with the control), and the expression of related apoptotic proteins in the mitochondrial pathway including cytochrome c and caspase-9 were up-regulated compared with the control (all P < 0.05). On the contrary, Bcl-2/Bax ratio was down-regulated compared with the control. Besides, the expression of pro-apoptotic proteins in the death receptor apoptosis pathway, including Fas-associating protein with a novel death domain and caspase-8, and the expression of caspase-3, a downstream regulatory protein of apoptosis, were also increased compared with the control (all P < 0.05). Inhibitors of caspase-9 and caspase-8 reversed the apoptosis process in H446 cells to varying degrees. Conclusions: These results suggest that iso-suillin could induce H446 cell apoptosis through the mitochondrial pathway and the death-receptor pathway. Therefore, iso-suillin might have a potential application as a novel drug for lung cancer treatment.
... To illustrate different modalities of cell death, we have used different cell lines: HT1080, L929, HaCaT, and HeLa cells (Feoktistova et al. 2011; Tenev et al. 2011). HaCaT cells (Boukamp et al. 1988) are primary keratinocyte-derived transformed cells that express low levels of cFLIP and XIAP, but that harbor high levels of cIAPs and caspases (Leverkus et al. 2000Leverkus et al. , 2003). HaCaT cells are highly sensitive to death receptor-induced cell death, and are thus a suitable model cell line to study molecular, biochemical , and morphological characteristics of the apoptotic and necroptotic signaling machinery. ...
Article
The processes by which cells die are as tightly regulated as those that govern cell growth and proliferation. Recent studies of the molecular pathways that regulate and execute cell death have uncovered a plethora of signaling cascades that lead to distinct modes of cell death, including “apoptosis,” “necrosis,” “autophagic cell death,” and “mitotic catastrophe.” Cells can readily switch from one form of death to another; therefore, it is vital to have the ability to monitor the form of death that cells are undergoing. A number of techniques are available that allow the detection of cell death and when combined with either knockdown approaches or inhibitors of specific signaling pathways, such as caspase or RIP kinase pathways, they allow the rapid dissection of divergent cell death pathways. However, techniques that reveal the end point of cell death cannot reconstruct the sequence of events that have led to death; therefore, they need to be complemented with methods that can distinguish all forms of cell death. Apoptotic cells frequently undergo secondary necrosis under in vitro culture conditions; therefore, novel methods relying on high-throughput time-lapse fluorescence video microscopy are necessary to provide temporal resolution to cell death events. Further, visualizing the assembly of multiprotein signaling hubs that can execute apoptosis or necroptosis helps to explore the underlying processes. Here we introduce a suite of techniques that reliably distinguish necrosis from apoptosis and secondary necrosis, and that enable investigation of signaling platforms capable of instructing apoptosis or necroptosis.
Article
Cancer is not a single disease but it constitutes a large variety of different types that are also different from each other phenotypically and molecularly. Although the standard treatments have resulted in clinical responses in a subset of patients, though, many patients relapse and no longer respond to further treatments. Hence, both the innate and adaptive resistance to treatments are the main challenges in today’s treatment strategies. Noteworthy, several novel treatment strategies, particularly immunotherapies, used alone or in combination, have been developed and that have significantly improved the therapeutic response of many unresponsive cancer patients. Nevertheless, even with the latest new developments of therapeutics that were effective in a larger subset of patients, there is still an urgent need to treat the remaining unresponsive subset of patients. This requires the development of new targeting agents of superior antitumor activities that will lead to overcoming the unaffected resistance by current treatments. There has been accumulating evidence suggesting nitric oxide donors as such targeting agents and considering their pleiotropic antitumor activities, including both the reversal of chemo and immuno-resistance of various unresponsive resistant cancers. The in vitro and in vivo preclinical findings corroborate the sensitizing antitumor activities of nitric oxide donors. In addition, a few clinical findings with NO donors that have been applied in patients have corroborated their antitumor and sensitizing activities in combination with standard therapies. In this review, the role and underlying mechanisms by which nitric oxide donors sensitize cancer resistant cells to both chemotherapy and immunotherapy are briefly described.
Article
Full-text available
The Bcl-2 family member Bcl-xLhas often been correlated with apoptosis resistance. We have shown recently that in peripheral human T cells resistance to CD95-mediated apoptosis is characterized by a lack of caspase-8 recruitment to the CD95 death-inducing signaling complex (DISC) and by increased expression of Bcl-xL (Peter, M. E., Kischkel, F. C., Scheuerpflug, C. G., Medema, J. P., Debatin, K.-M., and Krammer, P. H. (1997) Eur. J. Immunol. 27, 1207–1212). This raises the possibility that Bcl-xLdirectly prevents caspase-8 activation by the DISC. To test this hypothesis a cell line in which CD95 signaling was inhibited by overexpression of Bcl-xL was used. In these MCF7-Fas-bcl-xL cells Bcl-xL had no effect on the recruitment of caspase-8 to the DISC. It did not affect the activity of the DISC nor the generation of the caspase-8 active subunits p18 and p10. In contrast, cleavage of a typical substrate for caspase-3-like proteases, poly(ADP-ribose) polymerase, was inhibited in comparison with the control-transfected CD95-sensitive MCF7-Fas cells. To test whether Bcl-xL would inhibit active caspase-8 subunits in the cytoplasm, a number of immunoprecipitation experiments were performed. Using monoclonal antibodies directed against different domains of caspase-8, anti-Bcl-xLantibodies, or fusion proteins of glutathione S-transferase with different domains of caspase-8, no evidence for a direct or indirect physical interaction between caspase-8 and Bcl-xLwas found. Moreover, overexpression of Bcl-xL did not inhibit the activity of the caspase-8 active subunits p18/p10. Therefore, in this cell line that has become resistant to CD95-induced apoptosis due to overexpression of Bcl-xL, Bcl-xL acts independently and downstream of caspase-8.
Article
Full-text available
Caspase-3 is synthesized as a dormant proenzyme and is maintained in an inactive conformation by an Asp-Asp-Asp "safety-catch" regulatory tripeptide contained within a flexible loop near the large-subunit/small-subunit junction. Removal of this "safety catch" results in substantially enhanced autocatalytic maturation as well as increased vulnerability to proteolytic activation by upstream proteases in the apoptotic pathway such as caspase-9 and granzyme B. The safety catch functions through multiple ionic interactions that are disrupted by acidification, which occurs in the cytosol of cells during the early stages of apoptosis. We propose that the caspase-3 safety catch is a key regulatory checkpoint in the apoptotic cascade that regulates terminal events in the caspase cascade by modulating the triggering of caspase-3 activation.
Article
Death-inducing ligands (DILs) such as tumor necrosis factor α (TNFα) or the cytotoxic drug doxorubicin have been shown to activate a nuclear factor κB (NFκB)-dependent program that may rescue cells from apoptosis induction. We demonstrate here that TRAIL (TNF-related apoptosis-inducing ligand), a recently identified DIL, also activates NFκB in lymphoid cell lines in a kinetic similar to TNFα. NFκB activity is independent from FADD, caspases, and apoptosis induction. To study the influence of NFκB activity on apoptosis mediated by TRAIL, CD95, TNFα, or doxorubicin, NFκB activation was inhibited using the proteasome inhibitor N-acetyl-L-leucinyl-L-leucinyl-L-norleucinal or transient overexpression of mutant IκBα. Sensitivity for induction of apoptosis was markedly increased by these treatments in apoptosis sensitive cell lines. Moreover, both in cell lines and in primary leukemia cells that are resistant towards induction of apoptosis by DILs and doxorubicin, antagonization of NFκB activity partially restored apoptosis sensitivity. These data suggest that inhibition of NFκB activation may provide a molecular approach to increase apoptosis sensitivity in anticancer treatment.
Article
NF-κB, which consists of two polypeptides, p50 (M_r 50K) and p65/RelA (M_r 65K), is thought to be a key regulator of genes involved in responses to infection, inflammation and stress. Indeed, although developmentally normal, mice deficient in p50 display functional defects in immune responses. Here we describe the generation of mice deficient in the RelA subunit of NF-κB. Disruption of the relA locus leads to embryonic lethality at 15–16 days of gestation, concomitant with a massive degeneration of the liver by programmed cell death or apoptosis. Embryonic fibroblasts from RelA-deficient mice are defective in the tumour necrosis factor (TNF)-mediated induction of messenger RNAs for IκBα and granulocyte/macrophage colony stimulating factor (GM-CSF), although basal levels of these transcripts are unaltered. These results indicate that RelA controls inducible, but not basal, transcription in NF-κB-regulated pathways.
Article
Cytokines in the tumor necrosis factor (TNF) family regulate development and function of the immune system. We have isolated a new member of this family, designated Apo-2 ligand (Apo-2L), via an expressed sequence tag. Apo-2L is a 281-amino acid protein, related most closely to Fas/Apo-1 ligand. Transfected Apo-2L is expressed at the cell surface with its C terminus exposed, indicating a type II transmembrane protein topology. Like Fas/Apo-1 ligand and TNF, the C-terminal extracellular region of Apo-2L (amino acids 114-281) exhibits a homotrimeric subunit structure. Soluble Apo-2L induces extensive apoptosis in lymphoid as well as non-lymphoid tumor cell lines. The effect of Apo-2L is not inhibited by soluble Fas/Apo-1 and TNF receptors; moreover, expression of human Fas/Apo-1 in mouse fibroblasts, which confers sensitivity to induction of apoptosis by agonistic anti-Fas/Apo-1 antibody, does not confer sensitivity to Apo-2L. Hence, Apo-2L acts via a receptor which is distinct from Fas/Apo-1 and TNF receptors. These results suggest that, along with other family members such as Fas/Apo-1 ligand and TNF, Apo-2L may serve as an extracellular signal that triggers programmed cell death.
Article
TRAIL induces apoptosis through two closely related receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5). Here we show that TRAIL-R1 can associate with TRAIL-R2, suggesting that TRAIL may signal through heteroreceptor signaling complexes. Both TRAIL receptors bind the adaptor molecules FADD and TRADD, and both death signals are interrupted by a dominant negative form of FADD and by the FLICE-inhibitory protein FLIP. The recruitment of TRADD may explain the potent activation of NF-κB observed by TRAIL receptors. Thus, TRAIL receptors can signal both death and gene transcription, functions reminiscent of those of TNFR1 and TRAMP, two other members of the death receptor family.
Article
Apoptosis induced by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/APO-2L) has been shown to exert important functions during various immunological processes. The involvement of the death adaptor proteins FADD/MORT1, TRADD, and RIP and the apoptosis-initiating caspases-8 and -10 in death signaling by the two death-inducing TRAIL receptors 1 and 2 (TRAIL-R1 and TRAIL-R2) are controversial. Analysis of the native TRAIL death-inducing signaling complex (DISC) revealed ligand-dependent recruitment of FADD/MORT1 and caspase-8. Differential precipitation of ligand-stimulated TRAIL receptors demonstrated that FADD/MORT1 and caspase-8 were recruited to TRAIL-R1 and TRAIL-R2 independently of each other. FADD/MORT1- and caspase-8-deficient Jurkat cells expressing only TRAIL-R2 were resistant to TRAIL-induced apoptosis. Thus, FADD/MORT1 and caspase-8 are essential for apoptosis induction via TRAIL-R2.