Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 31, Issue of August 1, pp.

29184 –29191, 2003


© 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Necrotic Cell Death in Response to Oxidant Stress Involves the


Activation of the Apoptogenic Caspase-8/Bid Pathway*
Received for publication, February 14, 2003, and in revised form, May 14, 2003
Published, JBC Papers in Press, May 15, 2003, DOI 10.1074/jbc.M301624200

Xue Wang‡, Stefan W. Ryter‡, Chunsun Dai§, Zi-Lue Tang‡, Simon C. Watkins¶, Xiao-Ming Yin§,
Ruiping Song‡, and Augustine M. K. Choi‡储
From the ‡Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, the §Department of
Pathology, and the ¶Center for Biologic Imaging, Department of Cell Biology and Physiology, School of Medicine,
University of Pittsburgh, Pittsburgh, Pennsylvania 15213

Human epithelial (A549) cells exposed to hyperoxia biochemical, morphological, and functional characteristics (3).
die by cellular necrosis. In the current study, we dem- In necrosis, an extensive cell lysis results from acute, acciden-
onstrated the involvement of apoptogenic factors in ep- tal, or non-physiological injury (4). In contrast, apoptosis rep-
ithelial cell necrosis in response to hyperoxia, including resents a regulated form of cell death that participates in tissue
the formation of the Fas-related death-inducing signal- development and homeostasis, requiring the action of pro-
ing complex and initiation of mitochondria-dependent teases and nucleases within an intact plasma membrane (5).
apoptotic pathways. We showed increased activation of The delineation of apoptosis and necrosis as mutually exclusive
both Bid and Bax in A549 cells subjected to hyperoxia. processes, however, has not been convincingly demonstrated in
Bax activation involved a Bid-assisted conformational vivo or in vitro.
change. We discovered that the response to hyperoxia in Previous studies describe apoptosis as a major histological
vivo predominantly involved the activation of the Bid/
feature of hyperoxia-induced lung injury in vivo (6 – 8).
caspase-8 pathway without apparent increases in Bax
Petrache et al. (9) demonstrated induction of apoptosis in mu-
expression. Disruption of the Bid pathway by gene de-
rine macrophage cell lines in response to in vitro hyperoxia. In
letion protected against cell death in vivo and in vitro.
Likewise, inhibition of caspase-8 by Flip also protected contrast, hyperoxia primarily caused necrosis in A549 and in
against cell death. Taken together, we have demon- Type-I epithelial and murine lung bronchial cells (10 –12). Fur-
strated the involvement of apoptogenic factors in epi- thermore, pre-treatment with hyperoxia inhibited oxidant-in-
thelial cell responses to hyperoxia, despite a final out- duced apoptosis in A549 cells (13). Thus, the mechanisms un-
come of cellular necrosis. We have, for the first time, derlying hyperoxic lung injury and cell death may vary in a
identified a predominant role for the caspase-8/Bid tissue-specific manner and may involve apoptosis, necrosis, or
pathway in signaling associated with hyperoxic lung a mixed cell-death phenotype.
injury and cell death in vivo and in vitro. Members of the death receptor superfamily, including Fas
(14) and tumor necrosis factor-␣ (TNF-␣)1 (15), as well as Bcl
family members (i.e. Bax and Bcl-XL) have been implicated in
The clinical treatment of respiratory failure often requires hyperoxic lung injury. Adult mice exposed to hyperoxia (⬎95%
supplemental oxygen therapy. Unfortunately, exposure to an O2) for 72 h displayed increased whole-lung Bax and Bcl-XL
elevated oxygen tension (hyperoxia) may cause acute and mRNA levels; unaltered Bak, Bad, or Bcl-2 mRNA levels; and
chronic lung injury. Prolonged hyperoxia triggers an extensive decreased Bcl-w and Bfl-1 mRNA levels (14, 16). Increases in
inflammatory response in the lung that degrades the alveolar- Bcl-XL protein, but not Bax protein, have been reported in
capillary barrier, leading to impaired gas exchange and pulmo- response to hyperoxia in the mouse lung (16, 17). Hyperoxia
nary edema (1). The pathological changes in hyperoxia-injured may induce other cell death-related molecules, such as p53 and
lungs coincide with the injury or death of pulmonary capillary p21Cip1/WAF1/Sdi1 (p21) (14, 16, 18 –20). Expression of the p53
endothelial cells and alveolar epithelial cells, which maintain protein responds to DNA damage and, in turn, regulates genes
the integrity of the alveolar-capillary barrier and serve as a involved in growth control, DNA repair, and apoptosis. By
first-line defense against oxidative injury. Compromised epi- increasing the expression of pro-apoptotic Bcl-2 family mem-
thelial cell function may permit fluid and macromolecules to bers such as Bax, p53 may promote cell death. A major regu-
leak into the airspace, resulting in clinical respiratory failure latory target of p53, the cyclin-dependent kinase inhibitor
and death (2). Epithelial cell homeostasis requires stringent p21, may protect against oxidative lung injury by inhibiting
control over proliferative and apoptotic pathways. cell proliferation and DNA replication, and promoting DNA
Hyperoxia-induced cell death may involve apoptosis and ne- repair (11).
crosis, two distinct mechanisms of cell death with different Mice with different genetic backgrounds differ in oxygen
sensitivity. In vivo studies using genetically modified mouse
strains have revealed the relative roles of apoptosis-related
* This work was supported by National Institutes of Health Grants proteins in oxygen sensitivity of the lung. Fas⫺/⫺ (lpr) mice (14)
R01-HL60234, R01-AI42365, and R01-HL55330 (to A. M. K. C.). The
and TNFRI/II⫺/⫺ mice (21) did not display resistance to hyper-
costs of publication of this article were defrayed in part by the payment
of page charges. This article must therefore be hereby marked “adver-
tisement” in accordance with 18 U.S.C. Section 1734 solely to indicate
1
this fact. The abbreviations used are: TNF, tumor necrosis factor; TUNEL,
储 To whom correspondence should be addressed: Dept. of Medicine, Divi- terminal deoxynucleotidyl transferase-mediated dUTP nick end label-
sion of Pulmonary, Allergy, and Critical Care Medicine, 3459 Fifth Ave., ing; PBS, phosphate-buffered saline; LDH, lactate dehydrogenase;
Montefiore University Hospital, 628 NW, Pittsburgh, PA 15213. Tel.: 412- FasL, Fas ligand; DISC, death-inducing signal complex; TNFR-I, TNF
692-2210; Fax: 412-692-2260; E-mail: Choiam@msx.upmc.edu. receptor type I.
This is an Open Access article under the CC BY license.
29184 This paper is available on line at http://www.jbc.org
Pathways to Cell Death in Hyperoxia 29185
oxia. Murine embryonic fibroblasts derived from Bax⫺/⫺ and were incubated for 4 h at 37 °C. Then 2 ml of Dulbecco’s modified
Bak⫺/⫺ mice resisted hyperoxia-induced cytotoxicity (22). In Eagle’s medium containing 10% fetal bovine serum and antibiotics was
added to each plate. The plates were restored to the incubator and
contrast, p21⫺/⫺ mice were more sensitive than wild-type mice,
monitored every day until the fibroblasts reached confluence.
whereas p53⫺/⫺ mice did not display any modulation of oxygen Cross-linking of Fas—After treatment of hyperoxia, A549 cells were
sensitivity (11, 14, 16). Hyperoxia induced the expression of grown to 90% confluence and were then washed three times with cold
p21 and of growth arrest and DNA damage-inducible genes in PBS. Freshly prepared disuccinimyl suberate in Me2SO was then added
p53⫺/⫺ mice (11, 23). These observations suggest that down- to a final concentration of 0.25 mM to the cells in 1 ml of PBS. The dishes
stream regulatory targets of p53 such as p21 and Bax modulate were incubated for 30 min on a rocker platform. After cross-linking,
each dish was washed two times with PBS. The cells were then har-
cellular sensitivity to hyperoxia, although p53 itself is appar-
vested for Western blotting analysis.
ently not essential. Annexin V/Propidium Iodide Staining—Using the annexin V-FITC
Despite these observations, the one or more signal transduc- kit from BD Pharmingen we followed the manufacturer’s protocol.
tion pathways involved in hyperoxic lung injury and cell death Briefly, after hyperoxia treatment for 72 h, A549 cells were washed
remain incompletely understood. In the current study, we dem- with cold PBS and resuspended with binding buffer (10 mM HEPES/
onstrate the involvement of apoptogenic factors in epithelial NaOH, pH 7.4, 140 mM NaCl, 2.5 mM CaCl2) before transferring 1 ⫻ 105
cells to a 5-ml tube. Then 5 ␮l of annexin V and 5 ␮l of propidium iodide
cell responses to hyperoxia, despite a final outcome of cellular
were added, and the cells were incubated for 15 min in the dark.
necrosis. We have, for the first time, identified a predominant Binding buffer (400 ␮l) was then added to each tube and analyzed by
role for the caspase-8/Bid pathway in signaling associated with flow cytometry.
hyperoxic lung injury and cell death in vivo and in vitro. A DNA Ladder—A549 cells treated with hyperoxia were trypsinized
second pathway involving Bax gains importance under condi- and washed with PBS, and then the cells were spun down and resus-
tions that inhibit or abolish the caspase-8/Bid pathway. We pended in lysis buffer containing 50 mM Tris-HCl, pH 7.8, 10 mM
EDTA-2Na⫹, and 0.5% SDS. RNase A was then added at a concentra-
therefore provide an explanation for the lack of Bax modulation
tion of 0.5 mg/ml and incubated at 37 °C for 60 min. Next, the protein
in the lungs of wild-type mice subjected to a lethal dose of was degraded using 0.5 mg/ml proteinase K at 50 °C for 60 min. DNA
hyperoxia. fragmentation was visualized with ethidium bromide staining after
electrophoresis on a 1.2% agarose gel.
EXPERIMENTAL PROCEDURES Electron Microscopy Assays—Cell cultures were fixed in 2.5% glut-
Chemicals and Reagents—Digitonin, disuccinimidyl suberate, etopo- araldehyde in 0.1 M sodium cacodylate buffer (pH 7.2) for 1 h at 4 °C.
side, and other chemicals were from Sigma Chemical Co. (St. Louis, The cells were then postfixed in 1% osmium tetroxide, dehydrated in a
MO). Rabbit or goat polyclonal antibodies against Bax, Bcl-XL, Bid, graded series of ethanol, and embedded in LX112 (Ladd Corp.,
caspase-8, caspase-9, cytochrome c, Fas, TNFR-1, and protein A-aga- Burlington, VT). Thin sections (60 nm) were cut, stained with uranyl
rose were from Santa Cruz Biotechnology (Santa Cruz, CA) and were acetate and lead citrate, and examined on a Zeiss EM 10 transmission
used for Western blotting. Anti-Bax 6A7 antibody was purchased from electron microscope.
BD Pharmingen (San Diego, CA) and used for immunoprecipitation Cytosol Isolation—At different times after exposure to hyperoxia,
experiments. The Flip, Bcl-XL, and LacZ inserted in an adenovirus A549 cells were harvested in 0.05% of digitonin in extraction buffer
expression system were from the Molecular Medicine Institute Pro- containing 50 mM Hepes, pH 7.5, 50 mM KCl, 5 mM EGTA, and 2 mM
grams of Excellence in Gene Therapy Vector Core Facility, University of MgCl2 with protease inhibitors. The cell extracts were spun at 14,000 ⫻
Pittsburgh. g for 20 min, and the supernatants were removed and used for Western
Hyperoxia Exposure and Animal Experimentations—Normal, lpr blotting.
(Fas null, Fas⫺/⫺), and gld (Fas ligand null, FasL⫺/⫺) adult (7 weeks) Lactate Dehydrogenase Release Assay—LDH release was measured
pathogen-free male C3H/HeJ mice and appropriate controls were ob- using a commercially available assay (Cytotoxicity Detection Kit, Roche
tained from The Jackson Laboratory (Bar Harbor, ME). C57BL Bid Applied Science, Indianapolis, IN). Lung fibroblasts derived from the
wild-type (Bid⫹/⫹) and Bid-null (Bid⫺/⫺) adult (7 weeks) mice were bred wild-type or Bid⫺/⫺ strains were treated with hyperoxia. After gentle
at the University of Pittsburgh. Mice were kept in room air (control) or agitation, 200 ␮l of medium was removed at different time points to be
exposed to ⬎98% oxygen by placing the cages inside a plexiglass cham- used for the assay. The samples were incubated (30 min) with buffer
ber. Animals were injected with 1 ⫻ 108 of adenovirus (inserted with containing NAD⫹, lactate, and tetrazolium. LDH converts lactate to
appropriate gene) per mouse through the trachea into the lung 2 days pyruvate, thus generating NADH. The NADH then reduces tetrazolium
prior to oxygen exposure. Food and water were provided normally. (yellow) to formazan (red), which was detected by absorbance (490 nm).
Animals were observed closely for overall mortality, and the time of Statistical Analysis—Fisher’s exact test was used for analysis of the
death was recorded. Immediately after death, mice were necropsied, survival rate. The unpaired Student’s t test was used for other data
and their lungs were harvested for histochemical and biochemical analyses as indicated, and a value of p ⬍ 0.05 was considered
analyses. significant.
Cell Culture and Treatments—Human lung adenocarcinoma A549
RESULTS
cells (ATCC CCL185) were grown in F12K medium (Invitrogen) sup-
plemented with 10% fetal bovine serum. Cells were maintained at 37 °C Hyperoxia Induced Necrosis in A549 Epithelial Cells Involves
in 95% room air, 5% CO2 in a humidified chamber. Subconfluent cul- the Release of Apoptogenic Factors from the Mitochondria—
tures were used in all experiments, with the cells adhered 24 h prior to Morphological signs of injury appear in alveolar endothelial
the experimental treatment. Cells were cultured in sealed chambers
and epithelial cells within 48 –72 h of exposure to lethal levels
flushed with 95% O2 and 5% CO2. Control cells were cultured in 95%
room air, 5% CO2. For cells expressing the appropriate gene, 1 ⫻ 105 of of oxygen (2). Continued exposure for more than 72 h can result
adenovirus per milliliter was added to cells grown to about 30% conflu- in epithelial cell death, and consequently, loss of alveolar in-
ence into the medium 2 days before exposure to O2. Increased glucose tegrity, airway fluid accumulation, and mortality. Hyperoxia
consumption has been observed in a number of cell types exposed to primarily caused necrosis in A549 alveolar epithelial cells, as
hyperoxia in culture. In 10 ml of medium, glucose was depleted and determined by fluorescence DNA labeling, in situ end labeling
cellular ATP decreased after 36 h (24). We refreshed the medium and
of DNA, and electron microscopy (10).
gases daily to control for the equilibrium between apoptosis and necro-
sis that is influenced by maintaining the ATP levels (5). At each time We exposed human epithelial cells (A549) to in vitro hyper-
point, cell viability was determined by Trypan blue dye exclusion anal- oxia and then monitored the cells for survival. Flow cytometry
ysis (Invitrogen). Apoptosis was determined by morphological changes with PI/Annexin-V double staining and DNA-laddering analy-
and TUNEL assays. ses demonstrated that hyperoxia caused A549 cell death pri-
Preparation of Primary Fibroblast Cultures from Mouse Lung—Lung marily by necrosis (Fig. 1, B and C). Etoposide-induced apo-
tissue was excised from either wild-type or Bid⫺/⫺ mice. The lung tissue
ptosis of Jurkat cells was used as positive controls (Fig. 1A).
was soaked in a 1% antibiotic-antimycotic PBS (Invitrogen, Carlsbad,
CA) twice for 20 min. The tissue was minced into 1-mm pieces, which Quantitative data from our flow cytometry analyses (PI/An-
were plated onto p100 dishes (15–20 pieces per plate). Fetal bovine nexin-V double staining) demonstrate that ⬎96% of the dead
serum was added dropwise over each tissue piece, and then the plates cells were necrotic, whereas 3% were apoptotic after 72 h of
29186 Pathways to Cell Death in Hyperoxia

FIG. 1. Hyperoxia-induced necrosis in A549 cells. A, etoposide (100 ng/ml)-induced apoptosis of Jurkat cells for 6 h used as a positive control
for apoptosis. B and C, A549 cells were cultured under 95% room air/5% CO2 (Room air) or 95% O2/5% CO2 (hyperoxia) for the time indicated and
analyzed by PI/Annexin V double staining and flow cytometry (B) or DNA ladder analysis (C).

hyperoxia exposure. Furthermore, we also performed electron leads to the recruitment of procaspase-9 and its activation to
microscopy analysis of A549 cells at 72 h of hyperoxia, confirm- caspase-9.
ing the classic feature of necrosis as shown in Fig. 2. The The role of apoptotic pathways in hyperoxia-induced A549
epithelial cell growth medium was refreshed daily to maintain epithelial cell necrosis was evaluated in vitro, by monitoring
high levels of intracellular ATP (24). Hyperoxia induced the the expression of death receptors (Fas and TNFR-I) and Bcl-2
necrotic death of A549 cells even in the presence of a high family proteins. Following treatment of A549 cells with hyper-
glucose concentration in the medium. Under these experimen- oxia (24 –72 h), Fas protein levels decreased, whereas TNFR-I
tal conditions, ⬍50% of these cells survived after 72 h of hy- protein levels remained unchanged (Fig. 3B). Anti-Fas immu-
peroxia, by trypan blue analysis (Fig. 3A). noprecipitation revealed an interaction of caspase-8 with Fas
We hypothesized that apoptogenic factors may participate in (Fig. 3B), demonstrating DISC formation, the initiation of the
hyperoxia-induced A549 cell necrosis. In many cell types, apo- Fas-induced apoptotic pathway. We show here for the first
ptosis may proceed through receptor-dependent pathways ini- time, that hyperoxia up-regulated the activation of both Bax
tiated by Fas ligand (FasL) or tumor necrosis factor-␣ (TNF-␣). and Bid in A549 cells (Fig. 3C) with release of cytochrome c into
FasL-mediated apoptosis begins with the formation of a death- the cytosol (Fig. 3D). After 24 h exposure to hyperoxia, active
inducing signal complex (DISC), that involves the death recep- caspase-9 appeared in A549 cells (Fig. 3D).
tor Fas, Fas-associated death domain, and caspase-8. The con- We next examined the expression of the anti-apoptotic Bcl-2
sumption and recruitment of Fas leads to apparent decreases family member Bcl-XL as a function of hyperoxic exposure in
in Fas protein levels. Activated caspase-8 may in turn either A549 cells. Bcl-XL inhibits mitochondrial cytochrome c release
activate effector caspases (i.e. caspase-3) or cleave Bid. The and subsequent caspase activation (27–29). After 48 h in hy-
activated form of Bid triggers apoptosis by mitochondrial trans- peroxia, the expression of Bcl-XL increased in A549 cells (Fig.
location, which stimulates cytochrome c release (25). On the 3E), whereas Bcl-2 expression did not change (Fig. 3E). The
other hand, receptor-independent apoptosis may involve the high expression of Bcl-XL apparently did not protect A549 cells
mitochondrial-translocation of pro-apoptotic Bcl-2 family mem- from cell death.
bers such as Bax. In the mitochondria, Bax oligomers form Overexpression of FLIP Increased Survival under Hyperoxia
membrane channels by opening the permeability transition in Vitro and in Vivo—Because hyperoxia treatment led to
pore, leading to cytochrome c release and caspase activation caspase-8 and Bid activation, and increased Bcl-XL expression
(26). Once released in the cytosol, cytochrome c binds to Apaf-1, in A549 cells, we hypothesized that modulation of these apo-
stimulating its ATP-dependent oligomerization, which in turn ptosis-related proteins may influence oxygen sensitivity in
Pathways to Cell Death in Hyperoxia 29187
slightly decreased the expression of Bid protein and Bax con-
formational change relative to LacZ controls (Fig. 5). These
results support the hypothesis that Bid assists in a Bax con-
formational change that increases the efficiency of Bax in pro-
moting cell death.
lpr (Fas⫺/⫺) and gld (FasL⫺/⫺) Mice Did Not Display In-
creased Resistance to Hyperoxia but Displayed Increased Bax
Protein Expression Levels—We hypothesized that a Fas-in-
duced apoptosis-like process may precede necrotic cell death in
vivo. lpr mice (Fas⫺/⫺), however, did not display increased
resistance to hyperoxia (14). We challenged lpr (Fas⫺/⫺) and
gld (FasL⫺/⫺) mice with hyperoxia and monitored survival. As
shown in Fig. 6A, both Fas⫺/⫺ and FasL⫺/⫺ mice exhibited
similar survival curves in response to hyperoxia relative to the
C3H/HeJ wild-type mice, without significant statistical differ-
ence (p ⬎ 0.05) (Fig. 6A). We therefore investigated whether a
Fas-independent pathway, involving the expression of the
Bcl-2 family member Bax, might participate in the response to
hyperoxia. Under ambient conditions, Bax protein was ele-
vated in the lungs of the Fas⫺/⫺ mice relative to the wild-type,
with higher expression in Fas⫺/⫺ mice than in FasL⫺/⫺ mice
(Fig. 6B). Further increases in Bax protein were detected in the
lungs of Fas⫺/⫺ and FasL⫺/⫺ mice at the time of death by
hyperoxia, relative to hyperoxia-treated wild-type mice. Con-
versely, the activated form of Bid (p15) appeared only in the
lungs of wild-type mice subjected to hyperoxia (Fig. 6B). We
also found equivalent Bcl-XL expression and caspase-9 activa-
FIG. 2. Confirmation of necrosis in A549 cells induced by hy-
peroxia. A549 cells were cultured under 95% room air/5% CO2 (control)
tion in the lungs of all three mice strains after hyperoxia (Fig.
(A) and 95% O2/5% CO2 for 72 h (hyperoxia) (B) and analyzed by 6, B and C).
electron microscopy. These results suggest that the activation of Bid appears to
represent the dominant pathway leading to cell death in pul-
vitro. A549 cells were infected with adenoviral constructs in- monary cells during the hyperoxia response in vivo. Bid may
serted with Bcl-XL, Flip, or LacZ (Fig. 4A), exposed to in vitro assist in the conformational change of pre-existing Bax-protein,
hyperoxia for 72 h, and monitored for necrotic cell death. Over- which is not directly increased by hyperoxia in vivo. On the
expression of the caspase-8 inhibitor, FLIP (but not Bcl-XL), other hand, the deletion of the death receptor Fas promotes
significantly protected the cells from death during hyperoxia, increased Bax expression during in vivo hyperoxia, which
relative to the LacZ control (p ⬍ 0.05) (Fig. 4B). likely represents a compensatory activation of the Bax-induced
We hypothesized that inhibition of caspase-8 would also pathway.
modulate oxygen sensitivity in vivo. We infected C3H/HeJ mice Bid⫺/⫺ Mice Resisted Hyperoxia-induced Pulmonary Cell
with adenoviral constructs inserted with Flip, Bcl-XL, or LacZ. Death—We then hypothesized that deletion of apoptosis-re-
After 48 h, the mice were exposed to ⬎95% O2 and monitored lated factors downstream of Fas (i.e. Bid) would modulate ox-
for overall survival. Interestingly, the FLIP group was signifi- ygen sensitivity in vivo. Bid knock-out (Bid⫺/⫺) mice and cor-
cantly more resistant to oxygen toxicity than the LacZ-infected responding wild-type (Bid⫹/⫹) C57BL mice were exposed to
group (Fig. 4C). On the other hand, there was no significant ⬎95% O2 and monitored for overall survival and pulmonary
difference in hyperoxia resistance between the Bcl-XL-express- cell death. Interestingly, the Bid⫺/⫺ group was significantly
ing group and the LacZ control group (Fig. 4C). These results more resistant to oxygen toxicity than the corresponding wild-
emphasize an important role for the caspase-8/Bid pathway in type littermates (Fig. 7A). We also observed increased resist-
sensitivity to hyperoxia. These results also suggest that in- ance to hyperoxia in cells isolated from Bid⫺/⫺ null mice when
creases in Bcl-XL protein expression do not protect against compared with cells from negative littermate controls (Fig. 7B).
hyperoxia-induced lung injury or cell death. These results again emphasize an important role for the
Overexpression of FLIP in A549 Epithelial Cells Inhibited caspase-8/Bid pathway in sensitivity to hyperoxia.
Bid Expression, and Bax Conformational Change Induced by
Hyperoxia—Bid assists Bax in a conformational change that DISCUSSION
unmasks the Bax NH2-terminal domain and coincides with The therapeutic application of supplemental oxygen to ele-
mitochondrial cytochrome c release (30). Thus, the Bax mito- vate arterial pO2 and reduce tissue hypoxia can be offset by its
chondrial membrane insertion triggered by Bid may represent cytotoxic properties, which increase the risk of morbidity and
a key step in the pathways leading to apoptosis (31). Therefore mortality. Over the last 30 years, morphological studies in
we investigated the hypothesis that Bax conformational change animal models have described hyperoxia-induced lung injury.
may respond to the levels of active Bid in A549 cells subjected The first signs of damage include focal cytoplasm swelling of
to hyperoxia. A549 cells were infected with adenoviral con- microvascular endothelial cells, interstitial edema, and endo-
structs containing Bcl-XL, Flip, or LacZ insertions and then thelial cell fragmentation. After 72 h of exposure, type-I pul-
grown under hyperoxia (72 h). Cell lysates were immunopre- monary epithelial cells begin to die by necrosis (33–35). Many
cipitated with the anti-Bax monoclonal antibody (6A7) that studies have also demonstrated that lung cells exposed to hy-
specifically recognizes the conformational change in Bax pro- peroxia exhibit features of apoptosis, including chromatin con-
tein (32). FLIP overexpression decreased Bid protein levels and densation, DNA fragmentation, changes in the expression of
inhibited the conformational change of Bax, compared with Bcl-2 family genes, and increases in TUNEL-positive cells (12,
LacZ controls (Fig. 5). In contrast, Bcl-XL overexpression only 14, 16, 19, 20). We have also observed increases in TUNEL-
29188 Pathways to Cell Death in Hyperoxia

FIG. 3. Human lung epithelial cells, A549, are killed by hyperoxia, in association with the activation of apoptogenic factors. A, A549
cells were cultured in quadruplet under 95% room air/5% CO2 (0) and 95% O2/5% CO2 for the indicated hours. The extent of death was determined
by Trypan blue dye exclusion, and the data represent the average of three independent experiments (means ⫾ S.E.). The value for 72 h in hyperoxia
is significantly different (**, p ⬍ 0.01) from the control (0) or shorter hyperoxic exposures, using the unpaired Student’s t test. B–E, cell lysates were
then subjected to Western blot analyses as indicated to detect TNFR-1, Fas with/without cross-linking and caspase-8, (the latter from samples of
immunoprecipitation with anti-Fas) (B), Bax and Bid (C), cytochrome c and caspase-9 (D), and Bcl-2 and Bcl-XL (E).

positive staining in mouse lungs after hyperoxia treatment initiated by formation of an apoptosome complex, triggered by
relative to air-treated controls but without apparent differ- environmental insults, senescence, and developmental pro-
ences in the response between the various genetically modified grams, that involves the release of cytochrome c from the
strains or during overexpression of FLIP or Bcl-XL, as used in mitochondrial intermembrane space to the cytosol (5). The
this study (data not shown). Intravenous infusion of the Bcl-2 family of proteins controls mitochondrial integrity,
caspase inhibitor N-benzylcarbonyl-Val-Ala-Asp-fluoromethyl- whereby the balance of pro-apoptotic (Bax) and anti-apoptotic
ketone into mice, however, did not protect pulmonary cells from (Bcl-2 and Bcl-XL) proteins determines the relative sensitivity
death, nor affect lung weights and DNA laddering (14). These of cells to apoptotic stimuli (37). Bcl-XL protects the mitochon-
features of hyperoxic lung injury suggest that apoptosis and dria by inhibiting the release of cytochrome c and subsequent
necrosis may occur exclusively in the different lung cell types, procaspase activation, and therefore modulates both extrinsic
and in some cases, may occur concomitantly in the same cell and intrinsic apoptotic cell death. The existence of necrotic cell
type. death pathways regulated by an intrinsic death program dis-
The intracellular ATP concentration may represent a down- tinct from that of apoptosis has also been proposed (38).
stream signal capable of directing cells toward either type of Previous studies have suggested that human A549 epithelial
cell death, according to the hypothesis that high energy levels cells subjected to hyperoxia died primarily by necrosis (10). Our
are required for the execution of the apoptotic program, results (Fig. 1) support that hyperoxia induces necrotic cell
whereas they are dissipated during necrosis (36). A number of death in A549 cells. The outcome of necrosis did not strictly
additional cellular factors, independent of caspase activation, depend on depletion of cellular ATP levels, because we re-
may influence the decision between apoptosis and necrosis, freshed the medium daily to maintain high levels of glucose
including the duration of mitochondrial membrane pore open- and intracellular ATP (24). The induction of necrotic cell death
ing, oxidative stress, and Bcl-2 expression. in A549 cells activated an apoptotic process that involved the
Caspases play an indispensable role in regulating many of formation of the Fas-related DISC. We demonstrated the in-
the morphological and biochemical features of apoptosis. Two creased expression, activation, and mitochondrial transloca-
main pathways of procaspase activation have been proposed: (i) tion of both Bid and Bax in A549 cells subjected to hyperoxia.
extrinsic activation of initiator pro-caspases, triggered by a Bid activation by caspase-8 involved cleavage to the p15 form,
death receptor (i.e. Fas and TNF-R1)-initiated receptosome whereas Bax activation involved a Bid-assisted conformational
complex and (ii) intrinsic activation of initiator procaspases change. Activation of both Bax and Bid stimulated the release
Pathways to Cell Death in Hyperoxia 29189

FIG. 4. FLIP (but not Bcl-XL) increased survival under hyperoxia in vitro and in vivo. A549 cells infected with an adenovirus-inserted
gene, LacZ, Bcl-XL, or Flip, were cultured in quadruplet under the same conditions and experimental procedures. A, cell lysates were subjected
to Western blot analysis as indicated to detect the expression of Bcl-XL and FLIP. B, the value of survival of Flip-infected cells after 72 h under
hyperoxia is significantly different (*, p ⬍ 0.05) from control cells (LacZ-infected), but no differences occurred between Bcl-XL-infected and control
cells. C, mice were infected with an adenovirus-inserted gene, LacZ, Bcl-XL, or Flip. 48 h post-infection the mice were placed in a chamber with
⬎98% O2 and monitored for the time of death (n ⫽ 10 in each group). The mice infected with Flip displayed a significantly delayed mortality
compared with mice with LacZ and Bcl-XL (p ⫽ 0.02). There was no difference between the two groups of mice infected with LacZ and Bcl-XL.

protein has not changed in normal mouse lung in response to


hyperoxia (Fig. 6B). We demonstrated for the first time that
Bid activation occurs in normal mouse lung in response to
hyperoxia. We therefore propose that a Bid-dependent pathway
dominates over a Bax-dependent pathway in wild-type mice
subjected to hyperoxia, with Bid assisting in the conforma-
tional change of pre-existing Bax protein (Fig. 8). This model is
consistent with the lack of apparent modulation of Bax protein
in vivo during hyperoxia (Refs. 16 and 17 and Fig. 6B).
In our studies we also found that the Bid⫺/⫺ genotype sig-
nificantly conferred resistance to hyperoxia-induced mortality
in vivo and in vitro. Likewise, inhibition of caspase-8 by over-
expression of Flip also resulted in a marked hyperoxia-resist-
FIG. 5. Flip (but not Bcl-XL) expression during hyperoxia de- ant phenotype. Although caspase-8 activates Bid with the
creased the level of Bid protein and blocked the change in Bax
highest efficiency, other activating species may include
protein conformation. A549 cells infected with an adenovirus-in-
serted gene, LacZ, Bcl-XL, or Flip, were cultured in quadruplet under granzyme-B, and caspase-3 (39 – 41). The existence of alterna-
the same conditions and experimental procedures. Cell lysates were tive routes to Bid activation may explain the apparent lack of a
subjected to Western blot analysis as indicated to detect the expression hyperoxia resistance phenotype in Fas⫺/⫺ mice.
of Bid and Bax, including immunoprecipitation with anti-Bax 6A7 In the present study, we also found that the anti-apoptotic
antibody as indicated.
molecule, Bcl-XL, could not protect against hyperoxic lung in-
jury and cell death in vivo and in vitro (Fig. 4, B and C). Indeed
of mitochondrial cytochrome c and cleavage of caspase-9 (Fig. it has been reported that Bcl-XL does not prevent Bax-induced
8). The inhibition of caspase-8 protected epithelial cells from mitochondrial damage (42). Bax induces cell death via apopto-
hyperoxia-induced necrotic death. We conclude that the early sis in cells with a low level of Bcl-XL, whereas induces necrosis
events in hyperoxia-induced epithelial cell death involve the in cells with high expression of Bcl-XL (42). Meilhac et al. (43)
initiation of both receptor-mediated and mitochondria-depend- described a similar balance between apoptosis and necrosis
ent apoptotic pathways, despite a final outcome of cellular responding to Bcl-2 expression levels.
necrosis. This evidence suggests that the intracellular events leading
In mouse models, previous studies show that hyperoxia in- to apoptosis and necrosis can occur sequentially. In the initial
duces high levels of mRNA transcription of Bcl-XL and Bax, but step, there is a collapse of mitochondrial membrane potential
not Bax protein, in mouse lungs. Here, we confirm that Bax and loss of cellular glutathione. At this stage, pharmacologic
29190 Pathways to Cell Death in Hyperoxia

FIG. 6. FasLⴚ/ⴚ (gld) and Fasⴚ/ⴚ (lpr) mice could not manifest an increased resistance to hyperoxia compared with normal mice
(wt) but displayed changes in the expression of Bax and Bid. A, mice (either gld or lpr) were put in a chamber with ⬎98% O2 and monitored
for the time of death. No statistically significant differences in mortality occurred among the three groups (n ⫽ 12 in each group). The lysates from
four lungs of each group, in which the mice died at the same time in different groups, were subjected to Western blot analysis to detect Bax, Bid,
and Bcl-XL (B) or caspase-9 (C).

FIG. 8. Model of hyperoxia-induced cell death pathways in the


lung.

the morphological outcome can resemble apoptosis, necrosis, or


ⴚ/ⴚ
a mixed phenotype (45). Based on our observations, we reason
FIG. 7. Bid mice and fibroblasts with Bid null were resistant
that, in some cell types or in some conditions, the process of
to hyperoxia. A, mice (either wild-type or Bid⫺/⫺) were put in a
chamber with ⬎98% O2 and monitored for the time of death (n ⫽ 12 in necrosis shares a similar regulatory mechanism with apopto-
each group). There was a significant difference in mortality between the sis, which may include the activation of caspases. The final
two groups (p ⬍ 0.05). B, fibroblasts with/without Bid expression were manifestation of death is distinct from apoptosis, in that it
cultured in quadruplet under 95% room air/5% CO2 (0) and 95% O2/5% escapes inhibition by Bcl-XL.
CO2 for the indicated hours. LDH assay was used to test the death.
In summary, hyperoxia induces lung injury and leads to
necrotic cell death in lung epithelial cells, mainly through a two
intervention in the form of caspase inhibitors can prevent cell stage process: (i) the initiation of apoptosis-related molecules
death. In the second phase, a disruption of the plasma mem- leading to loss of normal mitochondrial function, which in-
brane leads to necrosis (44). Indeed, the term “programmed cell volves both death receptor-initiated (Bid-dependent) and mito-
death” now refers to any kind of cell death mediated by an chondria-dependent pathways leading to release of cytochrome
intracellular death program, irrespective of the trigger, where c and (ii) a necrotic cell death. The activation of Bid appears to
Pathways to Cell Death in Hyperoxia 29191
represent the dominant pathway in pulmonary cells during the 20. O’Reilly, M. A., Staversky, R. J., Watkins, R. H., and Maniscalco, W. M. (1998)
Am. J. Respir. Cell Mol. Biol. 19, 777–785
hyperoxia response in vivo. Bid assists Bax to change its con- 21. Pryhuber, G. S., O’Brien, D. P., Baggs, R., Phipps, R., Huyck, H., Sanz, I., and
formation into the active form (Fig. 8). These data, taken to- Nahm, M. H. (2000) Am. J. Physiol. Lung Cell Mol. Physiol. 278,
L1082–L1090
gether, suggest that cell death in the mouse lung and in cul- 22. Budinger, G. R., Tso, M., McClintock, D. S., Dean, D. A., Sznajder, J. I., and
tured human A549 epithelial cells involves a cellular death Chandel, N. S. (2002) J. Biol. Chem. 277, 15654 –15660
pathway sharing features of both apoptosis and necrosis. 23. O’Reilly, M. A., Staversky, R. J., Watkins, R. H., Maniscalco, W. M., and Keng,
P. C. (2001) Am. J. Physiol. 278, L552–L559
24. Allen, C. B., and White, C. W. (1998) Am. J. Physiol. 274, L159 –L164
REFERENCES
25. Yin, X. M. (2000) Cell Res. 10, 161–167
1. Horowitz, S., and Davis, J. M. (1997) in Lung Growth and Development, 26. Brady, H. J., and Gil-Gomez, G. (1998) Int. J. Biochem. Cell Biol. 30, 647– 650
(McDonald, J. A., ed) pp 577– 610, Decker Press, New York 27. Finucane, D. M., Bossy-Wetzel, E., Waterhouse, N. J., Cotter, T. G., and Green,
2. Mantell, L. L., and Lee, P. J. (2000) Mol. Genet. Metab. 71, 359 –370 D. R. (1999) J. Biol. Chem. 274, 2225–2233
3. Majno, G., and Joris, I. (1995) Am. J. Pathol. 146, 3–15 28. Susin, S. A., Lorenzo, H. K., Zamzami, N., Marzo, I., Snow, B. E., Brothers,
4. Alison, M. R., and Sarraf, C. E. (1994) Gut 35, 577–581 G. M., Mangion, J., Jacotot, E., Costantini, P., Loeffler, M., Larochette, N.,
5. Kroemer, G., Dallaporta, B., and Resche-Rigon, M. (1998) Annu. Rev. Physiol. Goodlett, D. R., Aebersold, R., Siderovski, D. P., Penninger, J. M., and
60, 619 – 642 Kroemer, G. (1999) Nature 397, 441– 446
6. Mantell, L. L., Horowitz, S., Davis, J. M., and Kazzaz, J. A. (1999) Ann. N. Y. 29. Wu, M., Xu, L., Li, X., Zhai, Z., and Shu, H. (2002) J. Biol. Chem. 277,
Acad. Sci. 887, 171–180 25617–25623
7. Otterbein, L. E., Chin, B. Y., Mantell, L. L., Stansberry, L., Horowitz, S., and 30. Desagher, S., Osen-Sand, A., Nichols, A., Eskes, R., Montessuit, S., Lauper, S.,
Choi, A. M. (1998) Am. J. Physiol. 275, L14 –L20 Maundrell, K., Antonsson, B., and Martinou, J. C. (1999) J. Cell Biol. 144,
8. Mantell, L. L, Shaffer, T. H., Horowitz, S., Foust, R., 3rd, Wolfson, M. R., Cox, 891–901
C., Khullar, P., Zakeri, Z., Lin, L., Kazzaz, J. A., Palaia, T., Scott, W., and 31. Eskes, R., Desagher, S., Antonsson, B., and Martinou, J. C. (2000) Mol. Cell.
Davis, J. M. (2002) Am. J. Physiol. 283, L31–L41 Biol. 20, 929 –935
9. Petrache, I., Choi, M. E., Otterbein, L. E., Chin, B. Y., Mantell, L. L., Horowitz, 32. Murphy, K. M., Streips, U. N., and Lock, R. B. (2000) J. Biol. Chem. 257,
S., and Choi, A. M. (1999) Am. J. Physiol. 277, L589 –L595 17225–17228
10. Kazzaz, J. A., Xu, J., Palaia, T. A., Mantell, L., Fein, A. M., and Horowitz, S. 33. Raffray, M., and Cohen, G. M. (1997) Pharmacol. Ther. 75, 153–177
(1996) J. Biol. Chem. 271, 15182–15186 34. O’Reilly, M. A. (2001) Am. J. Physiol. 281, L291–L305
11. O’Reilly, M. A., Staversky, R. J., Watkins, R. H., Reed, C. K., de Mesy Jensen, 35. Horowitz, S. (1999) Chest 116, Suppl. 1, 64S– 67S
K. L., Finkelstein, J. N., and Keng, P. C. (2001) Am. J. Respir. Cell Mol. 36. Leist, M., Single, B., Castoldi, A. F., Kuhnle, S., and Nicotera, P. (1997) J. Exp.
Biol. 24, 703–710 Med. 185, 1481–1486
12. McGrath-Morrow, S. A., and Stahl, J. (2001) Am. J. Respir. Cell Mol. Biol. 25, 37. Oltvai, Z. N., and Korsmeyer, S. J. (1994) Cell 79, 189 –192
150 –155 38. Kitanaka, C., and Kuchino, Y. (1999) Cell Death Differ. 6, 508 –515
13. Franek, W. R., Horowitz, S., Stansberry, L., Kazzaz, J. A., Koo, H. C., Li, Y., 39. Heibein, J. A., Goping, I. S., Barry, M., Pinkoski, M. J., Shore, G. C., Green,
Arita, Y., Davis, J. M., Mantell, A. S., Scott, W., and Mantell, L. L. (2001) D. R., and Bleakley, R. C. (2000) J. Exp. Med. 192, 1391–1402
J. Biol. Chem. 276, 569 –575 40. Li, H., Zhu, H., Xu, C. J., and Yuan, J. (1998) Cell 94, 491–501
14. Barazzone, C., Horowitz, S., Donati, Y. R., Rodriguez, I., and Piguet, P. F. 41. Luo, X., Budihardjo, I., Zou, H., Slaughter, C., and Wang, X. (1998) Cell 94,
(1998) Am. J. Respir. Cell Mol. Biol. 19, 573–581 481– 490
15. White, C. W., and Ghezzi, P. (1989) Biotherapy 1, 361–367 42. Hu, Y., Benedict, M. A., Wu, D., Inohara, N., and Nunez, G. (1998) Proc. Natl.
16. O’Reilly, M. A., Staversky, R. J., Huyck, H. L., Watkins, R. H., LoMonaco, Acad. Sci. U. S. A. 95, 4386 – 4391
M. B., D’Angio, C. T., Baggs, R. B., Maniscalco, W. M., and Pryhuber, G. S. 43. Meilhac, O., Escargueil-Blanc, I., Thiers, J. C., Salvayre, R., and Negre-
(2000) Lab. Invest. 80, 1845–1854 Salvayre, A. (1999) FASEB J. 13, 485– 494
17. Ward, N. S., Waxman, A. B., Homer, R. J., Mantell, L. L., Einarsson, O., Du, 44. Fujita, M., Kuwano, K., Kunitake, R., Hagimoto, N., Miyazaki, H., Kaneko, Y.,
Y., and Elias, J. A. (2000) Am. J. Respir. Cell Mol. Biol. 22, 535–542 Kawasaki, M., Maeyama, T., and Hara, N. (1998) Int. Arch. Allergy Immu-
18. McGrath, S. A. (1998) Am. J. Respir. Cell Mol. Biol. 18, 179 –187 nol. 117, 202–208
19. O’Reilly, M. A., Staversky, R. J., Stripp, B. R., and Finkelstein, J. N. (1998) 45. Denecker, G., Vercammen, D., Declercq, W., and Vandenabeele, P. (2001) Cell
Am. J. Respir. Cell Mol. Biol. 18, 43–50 Mol. Life Sci. 58, 356 –370

You might also like