Sei sulla pagina 1di 13

Journal of Neuroscience Research 75:83–95 (2004)

Oxidative Stress Induces p53-Mediated


Apoptosis in Glia: p53 Transcription-
Independent Way to Die
Paolo Bonini,1 Simona Cicconi,4 Alessio Cardinale,2 Cristiana Vitale,3
Anna Lucia Serafino,4 Maria Teresa Ciotti,5 and Lionel N.J-L. Marlier4*
1
Department of Internal Medicine, University of Rome Tor Vergata, Rome, Italy
2
Department of Neuroscience, University of Rome Tor Vergata, Rome, Italy
3
Department of Medical Science, San Raffaele Roma Hospital, Rome, Italy
4
Section of Molecular Medicine, Signal Transduction of Apoptotic Mechanisms Laboratory, National Research
Council (CNR), Institute of Neurobiology and Molecular Medicine (INeMM), Rome, Italy
5
Section of Neuroscience, National Research Council (CNR), Institute of Neurobiology and Molecular
Medicine (INeMM), Rome, Italy

Oxidative stress has been implicated in the pathogenesis mediator of stress response because it plays an essential role
of stroke, traumatic brain injuries, and neurodegenerative in the death of many cell types, other than neurons. It is
diseases affecting both neuronal and glial cells in the well known that p53 induces cell death by a multitude of
central nervous system (CNS). The tumor suppressor molecular pathways involving activation of target genes
protein p53 plays a pivotal function in neuronal apoptosis (i.e., pro-apoptotic Bax) (Miyashita and Reed, 1995; Jor-
triggered by oxidative stress. We investigated the role of dan et al., 1997; Brady et al., 1998; Xiang et al., 1998;
p53 and related molecular mechanisms that support ox- Morrison et al., 2003). Nevertheless, evidence for
idative stress-induced apoptosis in glia. For this purpose, transcription-independent pathways of p53-mediated ap-
we exposed C6 glioma cells and primary cultures of rat optosis is accumulating (Gottlieb and Oren, 1998; San-
cortical astrocytes to an H2O2-induced oxidative stress some et al., 2001), showing that p53 can contribute to
protocol followed by a recovery period. We evaluated the apoptosis by direct signaling at the mitochondria (March-
effects of pifithrin-␣ (PF-␣), which has been reported to enko et al., 2000) leading to cytochrome c (cyt-c) release
protect neurons from ischemic insult by specifically in- and caspase activation (Schuler et al., 2000).
hibiting p53 DNA-binding activity. Strikingly, PF-␣ was A key player in p53 regulation is MDM-2 protein.
unable to prevent oxidative stress-induced astrocyte ap-
MDM-2 can inhibit p53 binding within its transactivation
optosis. We demonstrate that p53 is able to mediate an
apoptotic response by direct signaling at mitochondria,
domain interfering with recruitment of basal transcription
despite its transcriptional activity. The z-VAD-fmk-
machinery components. Moreover, MDM-2 can lead to
sensitive apoptotic response requires a caspase- complete elimination of p53 through the ubiquitin-
dependent MDM-2 degradation, leading to p53 mito- proteasome pathway (Momand et al., 1992, 2000; Barak et
chondrial targeting accompanied by cytochrome c al., 1993; Wu et al., 1993; Zauberman et al., 1993; Chen et
release and nucleosomal fragmentation. al., 1996; Haupt et al., 1996). In addition, p53 binds specif-
© 2003 Wiley-Liss, Inc. ically to MDM-2 gene promoter establishing an important
autoregulatory feedback loop (Barak et al., 1993; Wu et al.,
Key words: astrocyte; C6 glioma; ischemia; MDM2; cy- 1993; Haupt et al., 1997; Kubbutat et al., 1997).
tochrome; mitochondria; caspase Recently, a small molecule was isolated for its ability to
reversibly block p53 transcriptional activity. This compound,

Apoptotic cell death plays an important role in brain Contract grant sponsor: San Raffaele Roma Hospital; Contract grant spon-
development as well as in neuronal injury and disease. It is sor: CNR-INeMM; Contract grant sponsor: the Italian Ministry of Uni-
versity Research, and Technology (MURST).
now generally accepted that massive neuronal death due to
oxidative stress is a common characteristic of brain *Correspondence to: Lionel N.J-L. Marlier, National Research Council,
whether in stroke (Culmsee et al., 2001b; Cheng et al., Institute of Neurobiology and Molecular Medicine (INeMM), Viale Marx
2003; Love, 2003) or in neurodegenerative diseases such as 15-43, 00137 Rome, Italy. E-mail: l.marlier@in.rm.cnr.it
Alzheimer’s disease (de la Monte et al., 1997, 1998), Received 10 January 2003; Revised 15 August 2003; Accepted 17 August
Parkinson’s disease (Jenner, 2003; Tatton et al., 2003), and 2003
traumatic brain injury (Napieralski et al., 1999). The p53
tumor suppressor protein has been proposed as a key

© 2003 Wiley-Liss, Inc.


84 Bonini et al.

called pifithrin-␣ (PF-␣), protects neurons from apoptosis RNA was reverse transcribed for 1 hr at 42°C using 200 U
induced by ischemic injury and excitotoxic insults. More- Moloney murine leukemia virus (MMLV) reverse transcriptase
over, PF-␣ protects mice from the lethal genotoxic stress (Invitrogen) in the presence of 2.5 ␮M random hexamers and
associated with anticancer treatment without promoting the 200 ␮M nucleotides (Amersham Pharmacia Biotech, MI, Italy)
formation of tumors (Komarov et al., 1999). Protection by in 20 ␮L final volume. Successively, 2 ␮L of each cDNA were
PF-␣ was correlated with decreased p53 DNA-binding ac- PCR-amplified using 2.5 U Platinum Taq (Invitrogen) in the
tivity, decreased expression of the p53 target gene Bax and presence of 15 pmol of specific primers as follows:
suppression of mitochondrial dysfunction and caspase activa-
tion (Culmsee et al., 2001a,b). ● Bcl-2 up: 5⬘-TAT AAG CTG TCA CAG AGG G;
The present study was undertaken to investigate the down: 5⬘-CTC CCC ACA CAC ATG ACC
molecular mechanisms that support oxidative stress- ● BclxL/S up: 5⬘-TCC CAG AAA GGA TAC AGC;
induced apoptosis in glial cells. For this purpose, we de- down: 5⬘-TCA CTT CCG ACT GAA GAG
signed an in vitro model of H2O2-induced oxidative stress ● Bad up: 5⬘-CCA GAT CCC AGA GTT TGA GC;
using both rat primary cultures of astrocytes and C6 rat down: 5⬘-CTT TCC CCA AAT TTC GAT CC
glioma cell line. We examined the role of p53 on the onset ● Bak up: 5⬘-TCC TGC CTC TCA GGG CAA; down:
of apoptosis and investigated whether PF-␣ was able to 5⬘-AGT GGT CAG GGA GGC ACT T
prevent glial cell death. ● Bax up: 5⬘-TTC ATC CAG GAT CGA GCA G;
down: 5⬘-TTC TTC CAG ATG GTG AGC G
MATERIALS AND METHODS ● Bid up: 5⬘-ACT CTG AGG TCA GCA ATG GC;
Ethics down: 5⬘-TAG GGA AGG ATG TCT TCA CCT C
Animals used for astrocyte preparations received humane ● p53 up: 5⬘-GCC ATC TAC AAG AAG TCA CA;
care according to the standard criteria regarding animal use in down: 5⬘-GTC TTC CAG CGT GAT GAT G
EEC. ● MDM-2 up: 5⬘-CCA ACA TGT CTG TGT CTA
CCG; down: 5⬘-ACA ATG TGC TGC TGC TTC TC
C6 Cell Line ● H4 up: 5⬘-GGT AAA GGG CTT GGG AAA G;
C6 cells were maintained in log-phase growth by regular down: 5⬘-TTA ACC ACC GAA GCC GTA G
passage using Dulbecco’s modified Eagle’s medium (DMEM) ● GAPDH up: 5⬘-ACC ACA GTC CAT GCC ATC
supplemented with 10% fetal bovine serum (FBS), 2 mM AC; down: 5⬘-TCC ACC ACC CTG TTG CTG TA
L-glutamine, 50 IU/ml penicillin, and 50 ␮g/ml streptomycin
(Invitrogen, Milan, Italy). Cells were subcultured twice a week To normalize the cDNA amount to be used during the PCR
and maintained at 37°C in an atmosphere of 95% humidified air amplification, pilot experiments were carried out using histone 4
and 5% CO2. (H4) specific primers (not shown). For all templates, 30 cycles were
used allowing us to consider the basal level as a real representation
Primary Cultures of Rat Astrocytes of relative expression levels of genes of interest.
Rat primary cultures of astrocytes were prepared from To verify whether after 24 hr reperfusion PF-␣ remained
cortex of embryonic Day 14 (E14) rat embryos as described able to block p53 transcriptional activity, RT-PCR analysis of
previously in detail (Mercanti et al., 1987). mRNA expression levels for H4 (as a housekeeping control
gene) and the glyceraldehyde-3-phosphate dehydrogenase gene
Oxidative Stress/Recovery Protocol (GAPDH) were carried out and the linear dynamic range ana-
To mimic oxidative stress cells were exposed to 100 ␮M lyzed by densitometry analysis using a computer-based imaging
H2O2/100␮M FeSO4 (Fenton reagent) in Hank’s balanced sa- system (Fluor-S; Biorad, Hercules, CA).
line solution (HBSS; Invitrogen) for 1 hr (the stress period) In all experiments, cDNAs were denatured initially for
followed by a recovery period consisting of replacing cells in 5 min followed by 30 cycles consisting of: 45 sec denaturation;
normal growth medium (Joseph et al., 1997; Richter-Landsberg 30 sec annealing; and 50 sec extension (3 min final extension)
and Vollgraf, 1998; Avshalumov and Rice, 2002). The basal using a Perkin Elmer 9700 thermal cycler (Perkin Elmer, Foster
point refers to the intact cells whereas the T0 time point refers City, CA).
to the end of the stress period/beginning of the recovery period.
The cells were preincubated 3 hr with zVAD-fmk Cell Survival Analysis
(20 ␮M final concentration; Biomol, Plymouth Meeting, PA) Astrocyte survival was assessed by counting the number of
and PF-␣ (100 nM final concentration; Calbiochem, San Diego, intact nuclei after lysing cells in detergent-containing solution as
CA) before the stress period and both zVAD-fmk and PF-␣ described initially by Soto and Sonnenschein (1985) and mod-
were then replaced during the recovery period. ified successively by Volonte et al. (1994). In all cases, 4 dishes
were scored in 4 independent experiments.
PCR Analysis
Total RNA was extracted using the TRIAzol reagent Immunofluorescence Studies
according to the manufacturer’s recommendation (Invitrogen) For immunofluorescence studies, both C6 cells and primary
and extracted RNA was used for reverse transcription- astrocytes were seeded in poly-D-lysine-coated coverslips. Cells
polymerase chain reaction (RT-PCR) analysis. Briefly, 1 ␮g were washed three times in phosphate-buffered saline (PBS) and
Astrocyte Apoptosis and p53 85

fixed in freshly prepared 4% paraformaldehyde-PBS for 10 min at basal apoptosis level, i.e., the assay of nucleosomes present in the
room temperature. Permeabilization was carried out in PBS/Triton culture immediately before stress (also termed basal).
X-100 (0.2%) for 5 min at room temperature. After three washes in Data are presented as means ⫾ SEM. Each experiment
PBS, cells were exposed to the appropriate dilution of primary was repeated at least 3 times using different batches of cells.
antibodies in incubation buffer (0.2 mg/ml bovine serum albumin Statistical analysis was carried out by one-way analysis of vari-
[BSA] in PBS) for 1 hr at room temperature. Cells were labeled for ance (ANOVA) followed by Bonferroni’s post-hoc comparison
p53, cyt-c, and MDM-2. After three washes in PBS, cells were test. Results were considered statistically significant when P
exposed to secondary antibody in the same buffer for 45 min at value was below 0.05.
room temperature. Preparations were mounted with 70% glycerol
in PBS. Samples were examined with a Leica fluorescence micro- RESULTS
scope coupled to a CCD camera, equipped with a 100⫻ oil Analysis of Cell Death, Nuclear Morphology, and
immersion lens. The dye Hoechst 33258 (Sigma-Aldrich, MI, Italy) Nucleosomal Formation Assay in Glial Cells After
was used at 0.5 ␮g/ml in PBS for 30 sec at room temperature Reactive Oxygen Species Exposure
before the cells were fixed. Primary antibodies used were anti-p53
(1:200) and anti-cyt-c (1:200) from Pharmingen (Becton Dickin- Cell survival was analyzed in primary cultures of rat
son, MI, Italy) and anti-MDM-2 (1:200) from Santa Cruz Biotech- astrocytes (Fig. 1A). This analysis demonstrated that HBSS
nology (Santa Cruz, CA). Secondary antibody was a Texas Red- exposure during the stress period did not induce cell death
conjugated anti-mouse IgG (1:300; Calbiochem). even after 24 hr recovery. On the contrary, cultures ex-
posed to reactive oxygen species (ROS) exhibited cell
Confocal Fluorescent Imaging death: after 6 hr in untouched control cultures, there was
For confocal microscopy analyses, primary astrocyte cultures 53.98 ⫾ 3.98% viable cells; after 12 hr, 45.43 ⫾ 7.41%;
were incubated for 30 min with MitoTracker Red 580 (200 nM and after 24 hr, 28.91% ⫾ 7.67. This demonstrated that
final concentration; Molecular Probes Europe BV, Poortgebouw, after 24 hr recovery, more than 70% of the cells were dead.
The Netherlands) at the end of the recovery period then fixed with Similar data were obtained in C6 glioma cells (not shown).
freshly prepared 2.5% glutaraldehyde in PBS (Sigma-Aldrich) for To determine whether cell death was due to apoptosis,
30 min at 4°C. Successively glutaraldehyde-induced autofluores- a time course analysis of nuclear morphology using Hoechst
cence was quenched by incubating cultures in sodium borohydride 33258 labeling (Fig. 1B) and a nucleosomal formation assay
(2 mg/ml for 3 ⫻ 10 min; Sigma-Aldrich). Cells were then (Fig. 1C) was carried out. Both C6 glioma cells and primary
permeabilized and processed for p53 immunofluorescence using a astrocytes exposed to HBSS during the stress period did not
fluorescein isothiocyanate (FITC)-conjugated anti-mouse antibody enter apoptosis, even after 24 hr recovery, as demonstrated by
(1:500; Sigma-Aldrich) as above. Fluorescently labeled preparations the absence of chromatin condensation and the lack of nu-
were observed using a Leica TCS 4D confocal laser scanning cleosomal enrichment (Fig. 1B,C). On the contrary, after
microscope supplemented with an argon/krypton laser and 12 hr recovery, apoptotic nuclei could be seen clearly in both
equipped with 40⫻ 1.00 – 0.5 and 100⫻ 1.3– 0.6 oil immersion cell cultures exposed to ROS, with a marked increase after
lenses. Confocal sections were taken at intervals of 0.5 ␮m. 24 hr reperfusion (Fig. 1B). The same results were confirmed
Excitation/emission wavelengths employed were 488 nm/510 nm, by increased nucleosomal formation (Fig. 1C): the enrich-
for FITC labeling, and 568 nm/590 nm, for MitoTracker dye. The ment factor increased significantly to 3.20 ⫾ 0.22 in C6 cells
acquisitions were recorded, employing pseudo-color representa- (P ⬍ 0.05 vs. HBSS) and 2.07 ⫾ 0.36 in primary cultures of
tion. astrocytes (P ⬍ 0.05 vs. HBSS) after 12 hr reperfusion. This
increase was much more pronounced after 24 hr of reperfu-
Nucleosomal Formation Assay sion, reaching 8.90 ⫾ 0.45 and 6.69 ⫾ 1.30, respectively
Both C6 cells and primary astrocytes were seeded in 24-well (P ⬍ 0.01 vs. HBSS in both cases).
plates. After 48 hr and 8–10 days, respectively, the culture medium
was removed and cells were exposed to the oxidative stress protocol Time Course Semiquantitative RT-PCR Analysis
detailed above. Control cells were incubated with HBSS only of Various Bcl-2 Family Members and p53-
during the stress period. After recovery, cells were lysed and nu- Regulated Genes
cleosomal formation was assayed by quantitative in vitro measure- To test for possible involvement of p53 transcriptional
ment of cytoplasmic histone-associated DNA fragments (Cell activity in the onset of oxidative stress-induced apoptosis and,
Death Detection ELISAPLUS; Roche, MI, Italy), according to the eventually, a relationship with Bcl-2 family member mRNA
manufacturer’s recommendations. expression levels, we carried out a time course semiquanti-
Results are expressed as the enrichment factor obtained tative RT-PCR analysis on several potential targets (Fig. 2).
according to the formula: This analysis demonstrated that in C6 glioma cells, Bcl-2,
Bcl-xL, Bax, and Bak mRNA expression remained almost
共关ODexperimental兴 ⫺ 关ODbackground兴兲/共关ODcontrol兴 ⫺ 关ODbackground兴兲 unchanged throughout the experiment. On the contrary,
Bcl-xS and Bid transcripts were upregulated already during
where the experimental point refers to the assay of nucleosomes the stress period and their expression levels remained signif-
present at a given time point during stress or recovery periods, icantly higher compared to baseline during the recovery
background consists of OD from ELISA plate dishes where period. In addition, Bad mRNA tended to increase during
cellular extracts were omitted, and the control point refers to the recovery but this increase was more evident for MDM-2 and
86 Bonini et al.

Genomic Effects of PF-␣ and Role in Apoptotic


Response
We monitored after 24 hr of recovery the genomic
effects of PF-␣ on p53 transcriptional activity. We carried
out a step-cycle RT-PCR analysis of mRNA expression
levels in C6 cells for H4 (as a housekeeping control gene)
and GAPDH was used as a positive control because it is
known to be regulated by p53 (Chen et al., 1999).
This experiment demonstrated that whereas the H4
housekeeping gene expression level remained unaffected
by PF-␣ treatment, the GAPDH expression level was
decreased in a dose-dependent manner (20 –500 nM; Fig.
3A). Similar results were obtained on primary astrocytes
(not shown). We next carried out a nucleosomal forma-
tion assay in parallel on C6 glioma cells and primary
cultures of astrocytes exposed or not exposed to PF-␣
treatment (100 nM) after 24 hr recovery (Fig. 3B). In both
experimental models, 1 hr oxidative stress followed by
24 hr recovery markedly induced nucleosomal formation.
This increase reached 8.56 ⫾ 0.45 in C6 cells and 7.33 ⫾
0.75 in primary cultures of glia.
In the presence of PF-␣, the enrichment factors in
C6 cells and primary cultures of astrocytes were 7.65 ⫾
0.65 and 6.54 ⫾ 0.60, respectively. No significant differ-
ences were noted cells treated with PF-␣ and untreated
cells were compared.
Immunofluorescence Analysis of p53, Cyt-c, and
MDM-2 Intracellular Distribution
Time-course immunofluorescence analysis of p53,
cyt-c, and MDM-2 protein expression combined with
Hoechst labeling of the nuclei showed that p53 was detect-
able almost exclusively in the nucleus until the end of the
stress period in both experimental models (Fig. 4, top). The
expression level of p53 increased progressively over the 24-hr
Fig. 1. A: Cell viability assay in primary astrocytes exposed to HBSS or recovery period, showing mainly a cytoplasmic localization
ROS for 1 hr and followed by a 6-, 12-, or 24-hr recovery period. of the protein in both experimental systems. The increase in
After ROS exposure, dead cells represent almost 50% of the population p53 expression level was more pronounced in C6 cells when
after 6 hr and reach more than 70% after 24 hr. B: C6 cells and primary compared to that in primary astrocytes.
astrocytes exposed to oxidative stress (0.1 mM FeSO4/H2O2 for 1 hr) Cyt-c distribution analysis in both experimental models
undergo apoptosis only during the recovery period. Both cell types (Fig. 4, center) evidenced a typical mitochondrial localization
were exposed to ROS (or HBSS alone as control) for 1 hr and then in intact cells after 1 hr stress; filamentous mitochondria were
allowed to recover for 12 and 24 hr. Hoechst staining shows that real discerned clearly. After 3– 6 hr recovery, mitochondrial struc-
apoptotic signs, such as nuclear shrinking and chromatin condensation, ture was altered profoundly, showing the typical apoptotic
can be observed only after ROS exposure and 24 hr of recovery. Rare
apoptotic nuclei were also observed in primary astrocytes after 12 hr of
organization known as megamitochondria (MG). After 12
recovery. Arrows point to apoptotic nuclei. C: Nucleosomal formation and 24 hr of recovery, changes in mitochondrial structure and
assay demonstrating severe DNA fragmentation in both experimental apoptotic changes of cells were evidenced clearly and prob-
models, particularly after 24 hr of recovery. *P ⬍ 0.05; **P ⬍ 0.01. ably reflected cyt-c release. Hoechst labeling also evidenced
chromatin condensation in both cell systems after 12–24 hr of
recovery.
p53 mRNA; the latter was upregulated transiently but sig- Time course immunofluorescence analysis of
nificantly during recovery. MDM-2 (Fig. 4, bottom), showed in both models a very
In primary cultures of astrocytes, similar results oc- low expression level in control cells and at the end of the
curred for all transcripts studied except Bcl-2, Bcl-xL, and stress period. After 3 hr of recovery, MDM-2 seemed
MDM-2. Indeed, during 24 hr of recovery, Bcl-2 expres- strongly upregulated, showing a marked nuclear localiza-
sion was reduced slightly compared to a progressive in- tion. In both experimental models, MDM-2 immunore-
crease in Bcl-xL expression. MDM-2 expression levels activity started to decrease after 6 hr of recovery and after
remained unchanged. 12 hr was almost undetectable.
Astrocyte Apoptosis and p53 87

Fig. 2. Semiquantitative PCR analysis of


p53 and Bcl-2 family member mRNA
expression levels. This analysis shows that
concomitantly with p53, several pro-
apoptotic genes such as Bad, Bid and
Bcl-xS are upregulated during oxidative
stress and recovery periods. A concomitant
slight decrease was observed in expression
levels of anti-apoptotic genes such as Bcl-2
and Bcl-xL.

Fig. 3. A: Step-cycle RT-PCR analysis of GAPDH and H4 histone.


PF-␣ treatment leads to a marked shift of the amplification curve,
compared to that of untreated cells, starting at a concentration of
20 nM. On the contrary, the H4 housekeeping gene profile is not
affected by PF-␣ treatment. B: Nucleosomal formation assay. Both cell
types were exposed to 1 hr ROS application followed by 24 hr of
recovery in the presence (ROS ⫹ PF-␣) or absence (ROS) of PF-␣.
Control cells were exposed to HBSS alone. It seems that PF-␣ does not
prevent DNA fragmentation.

Confocal Microscopy Analysis of p53 Intracellular labeling after MitoTracker labeling of mitochondria (Fig.
Distribution 5). For this purpose, primary astrocytes were exposed to
To document further the intracellular distribution of 1 hr oxidative stress followed by 6 or 24 hr of recovery.
p53, we carried out confocal microscopy analysis of p53 Confocal microscopy analysis evidenced a clear mitochon-
88 Bonini et al.

Fig. 4. Top: Immunofluorescence anal-


ysis of p53 during oxidative stress-
induced injury. In both C6 and primary
astrocytes, p53 is expressed at very low
levels under basal conditions but in-
creases during stress periods, showing a
nuclear localization. After 24 hr of re-
covery, p53 localization changes mark-
edly, evidencing a translocation to the
cytoplasm. Center: Immunofluores-
cence analysis of cyt-c during oxidative
stress-induced injury. Mitochondrial
structure remodeling and cyt-c release in
primary astrocytes and C6 cells exposed
to oxidative stress-induced injury. Un-
der basal conditions, mitochondria dis-
play a healthy morphology characterized
by filamentous structures. Within 3– 6 hr
of reperfusion, megamitochondria for-
mation is evident. Subsequently (12–24
hr of recovery), mitochondrial disrup-
tion and chromatin condensation can be
observed. Bottom: Immunofluores-
cence analysis of MDM-2 during oxida-
tive stress-induced injury. In both C6
and primary astrocytes, MDM-2 shows a
low expression level at the end of the
stress period and a marked increase is
detectable after 3 hr of recovery, show-
ing a strong nuclear localization. Subse-
quently, MDM-2-like immunoreactiv-
ity decreases (see the 6-hr recovery
point), becoming almost undetectable
after 12 hr of recovery until the end of
the experiment.

drial localization of p53 after ROS exposure compared to zVAD-fmk, but not PF-␣ inhibited MDM-2 degradation
that in HBSS treated cells. (Fig. 6, bottom). Conversely, a marked reduction of p53
protein level was observed in zVAD-fmk but not in PF-␣
Effects of zVAD-fmk and PF-␣ on p53 and treated cells (Fig. 6, top).
MDM-2 Distribution After 6 Hours of Recovery
The p53 regulator MDM-2 contains a caspase-3-like Effects of zVAD-fmk on Nucleosomal Formation
cleavage site (Chen et al., 1997). We therefore wanted to Because caspase inhibition was able to modulate
monitor whether zVAD-fmk treatment could modulate both p53 and MDM-2 expression levels, we investi-
MDM-2 expression in our experimental paradigm. For gated whether this could inhibit apoptosis. We moni-
this purpose, C6 cells and primary astrocytes were exposed tored the effects of zVAD-fmk on nucleosomal forma-
to 1 hr of oxidative stress followed by 6 hr of recovery in tion in both experimental models after 24 hr recovery
presence or absence of the broad-spectrum caspase inhib- (Fig. 7). This assay demonstrated that in both C6 cells
itors zVAD-fmk or PF-␣. In both experimental models, and primary astrocytes, zVAD-fmk protected cells from
Astrocyte Apoptosis and p53 89

Fig. 5. Confocal microscopy analysis of


p53 redistribution during oxidative
stress-induced injury in primary cultures
of astrocytes after mitochondrial staining
(MitoTracker). HBSS control is pre-
sented on the first column after 24 hr of
recovery. Second and third column
show primary astrocytes exposed to 1 hr
ROS, after 6 and 24 hr recovery, respec-
tively. Immunolabeling of p53 is barely
detectable in HBSS-treated cells com-
pared to that in ROS-exposed astro-
cytes, where p53 can be seen in the
nucleus after 6 hr of recovery. Please
note on overlay lane the almost com-
plete overlay for MitoTracker and p53
labeling in ROS-treated cells, docu-
menting the p53 mitochondrial locali-
zation.

oxidative stress-induced apoptosis: the enrichment fac- DISCUSSION


tor decreased from 7.54 ⫾ 0.70 to 2.93 ⫾ 0.35 (P ⬍
Glial Cells Enter Apoptosis During Recovery After
0.01 vs. HBSS) in C6 cells and from 5.26 ⫾ 0.65 to ROS Exposure
1.84 ⫾ 0.75 (P ⬍ 0.01 vs. HBSS) in primary astrocytes
after 24 hr of recovery. To mimic oxidative stress injury, cultured rat cortical
astrocytes and C6 glioma cells were exposed to ROS in
Effects of zVAD-fmk and PF-␣ on Cyt-c HBSS for 1 hr followed by a recovery period in which cells
were placed in complete growth medium. Time course
Distribution After 24 Hours of Recovery
analysis of cell viability, nuclear morphology, (Hoechst 33258
To document further the effects of caspase blockade labeling) and a nucleosomal formation assay demonstrated
on apoptosis with respect to PF-␣, cyt-c immunolocaliza- that both C6 glioma cells and primary astrocytes die when
tion was monitored in both C6 cells and primary astro- exposed to ROS but not when exposed to HBSS during the
cytes exposed to 1 hr of oxidative stress followed by 24 hr stress period. Cell death involved up to 70% of the cells
recovery, treated or not treated with zVAD-fmk and within the 24 hr of recovery and was identified clearly as
compared to those treated with PF-␣ (Fig. 8). This analysis apoptotic cell death based on nuclear morphology after
evidenced that mitochondrial disruption, cyt-c release, Hoechst staining and enrichment of nucleosome formation.
and chromatin condensation were completely abrogated After 12 hr of recovery, apoptotic nuclei could be seen clearly
by zVAD-fmk treatment, whereas PF-␣ treatment did not in both cell cultures exposed to ROS, with a marked increase
impede these events. after 24 hr of recovery. This induction of apoptosis due to
90 Bonini et al.

Fig. 6. Immunofluorescence analysis of


p53 and MDM-2: effects of zVAD-fmk
and PF-␣ treatments. C6 and primary
astrocytes were exposed to ROS fol-
lowed by 6 hr of recovery (second col-
umn) compared to cells exposed to ox-
idative stress but treated with zVAD-fmk
(third column) or PF-␣ (fourth column).
Basal (first column) refers to intact cells.
MDM-2 degradation is inhibited by
zVAD-fmk, leading to concomitant p53
degradation. No p53 immunoreactivity
can be detected in cytosol of cells treated
with zVAD-fmk. PF-␣ did not interfere
with MDM-2 distribution or with p53
mitochondria-like accumulation.

ROS exposure was confirmed by the increase in nucleoso- Semiquantitative RT-PCR analysis (Fig. 2) showed
mal formation. that in both experimental models p53 mRNA was upregu-
Expression Levels of Bcl-2 Family Member lated transiently but significantly during recovery with a
mRNA Are Modulated During Recovery: Possible parallel upregulation of pro-apoptotic Bax and Bad genes.
Involvement of p53 Moreover, two other pro-apoptotic members of the Bcl-2
In recent years, a critical role for mitochondria in family, Bcl-xS and Bid, were upregulated already during the
apoptosis induction has been demonstrated (Matsuyama oxidative period. The anti-apoptotic genes Bcl-2 and Bcl-xL
and Reed, 2000). In particular, these organelles release remained almost unchanged in C6 glioma cells whereas in
proteins into the cytosol such as cyt-c and apoptosis- primary astrocytes, during 24 hr of recovery, Bcl-2 expres-
inducing factor (AIF), triggering caspase-dependent and sion was reduced slightly compared to a progressive increase
independent apoptotic mechanisms (Green, 1998; Reed et in Bcl-xL expression. Finally, MDM-2 mRNA was upregu-
al., 1998; Susin et al., 1998, 1999). Several pro-apoptotic lated in C6 cells, but not in primary astrocytes, during re-
proteins of Bcl-2 family, such as Bid, Bak, and Bax play a covery. The expression profile of pro-apoptotic genes ob-
pivotal role in this process (Eskes et al., 1998, 2000). In served in our system was in agreement with a previous
addition, expression of many members of the Bcl-2 pro- large-scale expression analysis of nonneuronal models of p53-
teins family are regulated by p53 (Kannan et al., 2000). induced apoptosis (Polyak et al., 1997).
Astrocyte Apoptosis and p53 91

Interestingly, although most pro-apoptotic genes ance of pro-apoptotic versus anti-apoptotic genes towards
were upregulated during oxidative stress simulation (T0 pro-apoptotic genes.
time point, Fig. 2), cells exhibited real apoptotic signs (i.e.,
nuclear condensation) only after several hours of recovery PF-␣ Inhibits p53 Transcriptional Activity but
(Fig. 1A). These data confirm observations published pre- Does Not Protect Glial Cells Against Oxidative
Stress-Induced Apoptosis
viously regarding p53 induction in the human glioblas-
toma cell line A172 after exposure to oxidative stress; Recent preclinical studies have demonstrated the
however, these authors reported an increase in Bak ex- efficacy of a p53 inhibitor in models of stroke and neuro-
pression, which did not occur in our system (Kitamura et degenerative disorders, and have suggested that drugs that
al., 1999). inhibit p53 may reduce the extent of brain damage in
All these findings suggest a possible role for p53 related human neurodegenerative conditions (Culmsee et
transcriptional activity on apoptosis onset, leading the bal- al., 2001a,b; Lakkaraju et al., 2001; Chung et al., 2002).
Based on these data, we tested the potential protective
properties of PF-␣.
We first carried out a pilot experiment using the
nucleosomal formation assay, to evaluate a potential pro-
apoptotic effect of PF-␣ treatment. We showed that, as
reported previously in neurons (Culmsee et al., 2001b),
PF-␣ was without significant effect on glial cell survival at
concentrations up to 200 nM, although higher concentra-
tions slightly induced apoptosis, as revealed by nucleoso-
mal formation assay (data not shown).
To verify whether after 24 hr of recovery PF-␣
remained able to block p53 transcriptional activity, we
used step-cycle RT-PCR analysis to monitor in C6 cells
the mRNA expression levels for H4, as a housekeeping
control gene, and GAPDH, because its expression is mod-
ulated by p53 as mentioned previously (Chen et al., 1999).
The dose-dependent decrease in GAPDH expression
compared to the lack of H4 housekeeping gene expression
modulation confirmed PF-␣ efficacy at THE genomic
Fig. 7. Nucleosomal formation assay in zVAD-fmk treated cultures. level (Fig. 3A).
Both cell types were exposed to 1 hr oxidative stress followed by 24 hr We therefore decided to use 100 nM PF-␣ to test its
of recovery in the presence (ROS ⫹ zVAD-fmk) or absence (ROS) of putative protective effect against oxidative stress-induced
zVAD-fmk. Control cells were exposed to HBSS alone. Nucleosomal apoptosis, in both rat primary cultures of cortical astrocytes
formation was prevented significantly by zVAD-fmk. and C6 glioma cells.

Fig. 8. Immunofluorescence analysis of


cyt-c: effects of zVAD-fmk and PF-␣
treatment. C6 cells and primary astro-
cytes were exposed to ROS followed by
24 hr of recovery (second column) com-
pared to cells exposed to oxidative stress
but treated with zVAD-fmk (third col-
umn) or PF-␣ (fourth column). Basal
(first column) refers to intact cells. In
both primary astrocytes and C6 cells
treated with zVAD-fmk (third column),
mitochondrial structure is preserved,
cyt-c immunoreactivity is not altered,
and chromatin condensation cannot be
observed. In cells treated with PF-␣
(fourth column), no peculiar difference
can be observed when compared to cells
exposed to oxidative stress (second col-
umn).
92 Bonini et al.

We analyzed nucleosomal formation in both exper- agreement with previous findings, these data suggested
imental cell models after 24 hr of recovery in the presence that p53 was able to induce apoptosis through its translo-
or absence of PF-␣ (Fig. 3B). Strikingly, we observed that cation to mitochondria and the subsequent release of cyt-c
PF-␣ failed to protect both primary astrocytes and C6 cells (Schuler et al., 2000).
against oxidative stress-induced apoptosis, despite its abil-
ity to inhibit p53 genomic effects. This was an unexpected Mitochondrial Localization of p53 Requires
result because in neurons, this compound was able to MDM-2 Degradation via Caspase Activity
prevent apoptosis induced by ischemic insult, as reported MDM-2 protein is a key player in the regulation of
previously (Komarov et al., 1999). In addition, the lack of p53. Thus, MDM-2 binds to p53 within its transactivation
protection against apoptosis after PF-␣ treatment further domain, interfering with recruitment of basal transcription
supports the hypothesis that discrepancies in various gene machinery components, and within the nucleus MDM-2
expression noticed between C6 cells and primary astro- can bind p53 targeting newly formed complexes toward
cytes may not have a primary importance for cell survival. the cytoplasm due to a nuclear export sequence (NES)
Intracellular Redistribution of p53 Toward present in the MDM-2 protein sequence. This leads to the
Mitochondria After Oxidative Stress Injury degradation of p53 by the ubiquitin-proteasome pathway
Nuclear bypass experiments demonstrated that mi- (Haupt et al., 1997; Kubbutat et al., 1997).
tochondrial localization of p53 is sufficient to induce p53- To investigate the involvement of MDM-2 in the
dependent apoptosis, indicating dual action of this protein apoptotic response in our experimental model, we carried
during apoptosis (Ding and Fisher, 1998; Ding et al., 1998; out a time course immunofluorescence analysis. As shown
Marchenko et al., 2000). in Figure 4, MDM-2 was expressed in both C6 and
Time-course immunofluorescence analysis of p53 primary cultures of rat astrocytes at very low levels until
protein combined with Hoechst labeling of the nuclei the end of ROS exposure, whereas p53 was upregulated
(Fig. 4), showed that p53 was detectable in both experi- already. MDM-2 reached its maximum expression level
mental models almost exclusively in the nucleus until the after 3 hr of recovery with a marked nuclear localization
end of the stress period. The p53 expression level increased and seemed barely detectable after 6 hr of recovery. Si-
progressively over the 24-hr recovery period, showing an multaneously, p53 immunoreactivity increased strongly,
almost cytoplasmic localization of the protein in both showing a marked cytosolic signal. After 12 hr of recovery
experimental systems. The p53 expression level seemed MDM-2 immunoreactivity decreased strongly becoming
higher in C6 when compared to that in primary astrocytes, almost undetectable until the end of the recovery period.
in agreement with RT-PCR analysis. These data suggested a direct relationship between the
It has been reported that when present in cytoplasm, increase in cytoplasmic p53 expression, and its mitochon-
most p53 protein is localized within the mitochondrial drial redistribution, and the corresponding MDM-2 ex-
membrane compartment, although a small fraction of the pression levels.
protein can be found in other organelles. This mitochon- MDM-2 contains a caspase-3-like cleavage site that
drial localization of p53 was confirmed by confocal mi- is evolutionarily conserved (Chen et al., 1997), and in
croscopy analysis in primary astrocytes (Fig. 5). vitro experiments carried out in H1299 cells, an epithelial
At the mitochondrial level, p53 exerts its pro- cell line expressing a temperature-sensitive human p53,
apoptotic function by favoring cyt-c release and subse- demonstrated that caspase-3 is able to cleave MDM-2.
quent caspase activation (Marchenko et al., 2000; Sansome The cleaved MDM-2 loses the ability to promote p53
et al., 2001; Schuler et al., 2000; Schuler and Green,
2001). To investigate further such a mechanism, we car- degradation and potentially functions in a dominant-
ried out a time course immunofluorescence analysis of negative fashion to stabilize p53 (Pochampally et al.,
cyt-c distribution. Both C6 cells and primary astrocytes 1999).
were exposed to ROS for 1 hr and analyzed within 24 hr To verify a possible relationship between MDM-2
of recovery. As shown in Figure 4 in control cells (basal degradation and p53 expression levels, we exposed both
point), filamentous mitochondria were discerned clearly C6 cells and primary astrocytes to oxidative stress injury in
whereas between 3– 6 hr of recovery and concomitantly the presence or absence of the broad-spectrum caspase
with p53 redistribution toward mitochondria (see Fig. 4 at inhibitor zVAD-fmk. We also analyzed the effect of PF-␣
6 hr recovery), the mitochondrial structure was altered treatment. We demonstrated that p53 and MDM-2 ex-
profoundly, showing a typical apoptotic organization pression levels displayed a complementary but opposite
known as megamitochondria (MG). At this point, cyt-c profile after 6 hr of recovery (Fig. 4). As shown in Figure
release was not yet detectable, leading to a situation that 6, in both C6 cells and primary astrocytes, zVAD-fmk, but
usually, but not necessarily, precedes apoptosis (Karbowski not PF-␣ inhibited MDM-2 degradation, leading to a
et al., 1999; Teranishi et al., 2000). After 24 hr recovery, marked reduction of p53 protein level and prevention of
changes in mitochondrial structure and apoptotic changes its translocation toward mitochondria.
in cells were marked and showed the swelling of free These findings suggest a cause-and-effect relation-
radical-induced megamitochondria, which in turn were ship between caspase activity, MDM-2 degradation, and
responsible for cyt-c release and nuclear shrinking. In p53 protein stabilization.
Astrocyte Apoptosis and p53 93

Fig. 9. Proposed mechanism of oxida-


tive stress-induced apoptosis in glia cells.
Dashed arrows represent the physiolog-
ical pathway in which p53 contributes to
MDM-2 transcription. When exported
into the cytoplasm, MDM-2 protein
binds p53, driving its degradation
through ubiquitin-proteasome pathway.
This contributes to maintain low p53
expression level. In the presence of ROS
(large arrows), partial mitochondrial
membrane depolarization leads to cyt-c
release and caspase activation, which in
turn contributes to MDM-2 degrada-
tion. p53 can translocate toward mito-
chondria, leading to a massive cyt-c re-
lease and establishing an apoptotic loop.
Treatment with zVAD-fmk abolishes
onset of the apoptotic loop. PF-␣ is un-
able to protect cells from apoptosis, in-
dicating that p53 transcriptional activity
is not the main pathway involved in our
model of oxidative stress-induced apo-
ptosis.

Pro-apoptotic Mitochondrial Function of p53 Is cell death. For this purpose, we monitored nucleosomal
Abrogated Completely by zVAD-fmk but not by formation in both experimental models after to 24 hr of
PF-␣ recovery in the presence or absence of zVAD-fmk (Fig. 7).
We demonstrated that, in our models, p53 redistri- This assay demonstrated that in both C6 cells and primary
bution within the cytosol is related to cyt-c release from astrocytes, zVAD-fmk protected cells from ROS
mitochondria and cell death execution (Fig. 4). In the exposure-induced apoptosis; indeed, the enrichment fac-
presence of zVAD-fmk, p53 mitochondrial targeting was tor was decreased strongly in both cell cultures after 24 hr
abrogated completely by preventing MDM-2 caspase- of recovery.
dependent degradation. The question then raised was Finally, we investigated whether zVAD-fmk protec-
whether this process was sufficient to prevent apoptotic tion was related to the inhibition of p53-mediated cyt-c
94 Bonini et al.

release. Both C6 cells and primary astrocytes were exposed (to P.B., S.C., and A.C. from Ministry of Education,
to ROS, treated or not with zVAD-fmk and compared to University and Research, Miur-Italy).
sisters cultures exposed to ROS not treated with PF-␣.
This analysis evidenced that after 24 hr of recovery, no REFERENCES
cyt-c released could be observed and mitochondria dis- Avshalumov MV, Rice ME. 2002. NMDA receptor activation mediates
played a filamentous and healthy morphology in zVAD- hydrogen peroxide-induced pathophysiology in rat hippocampal slices.
fmk treated cells only. J Neurophysiol 87:2896 –2903.
In conclusion, we have found that in a glial model of Barak Y, Juven T, Haffner R, Oren M. 1993. mdm2 expression is induced
oxidative stress-induced apoptosis, p53 plays a pivotal role by wild type p53 activity. EMBO J 12:461– 468.
by directly signaling mitochondria despite its transcrip- Brady HJ, Gil-Gomez G. 1998. Bax. The pro-apoptotic Bcl-2 family
tional activity. PF-␣ fails to protect glial cells from ROS member, Bax. Int J Biochem Cell Biol 30:647– 650.
Chen J, Wu X, Lin J, Levine AJ. 1996. mdm-2 inhibits the G1 arrest and
exposure despite its ability to inhibit p53 genomic effects, apoptosis functions of the p53 tumor suppressor protein. Mol Cell Biol
contrary to what has been observed in neuronal cells 16:2445–2452.
(Komarov et al., 1999). Our data suggest that in the Chen L, Marechal V, Moreau J, Levine AJ, Chen J. 1997. Proteolytic
apoptotic cascade, three fundamental steps are involved. cleavage of the mdm2 oncoprotein during apoptosis. J Biol Chem 272:
The first step, engagement, involves low cyt-c release from 22966 –22973.
mitochondria within first 3 hr of reperfusion (probably Chen RW, Saunders PA, Wei H, Li Z, Seth P, Chuang DM. 1999.
due to a partial mitochondrial membrane depolarization, Involvement of glyceraldehyde-3-phosphate dehydrogenase (GAPDH)
as evidenced by JC-1 mitochondrial potential sensor anal- and p53 in neuronal apoptosis: evidence that GAPDH is upregulated by
ysis; data not shown). This is followed by partial caspase p53. J Neurosci 19:9654 –9662.
activation, which in turn acts on early targets such as Cheng T, Liu D, Griffin JH, Fernandez JA, Castellino F, Rosen ED,
MDM-2, leading to a redistribution of p53 toward mito- Fukudome K, Zlokovic BV. 2003. Activated protein C blocks p53-
mediated apoptosis in ischemic human brain endothelium and is neuro-
chondria. In the second step, commitment, concomitantly protective. Nat Med 9:338 –342.
with p53 redistribution into the cytosol, mitochondria Cheng Y, Deshmukh M, D’Costa A, DeMaro JA, Gidday JM, Shah A, Sun
modify their structure by forming megamitochondria Y, Jacquin MF, Johnson EM, Holtzman DM. 1998. Caspase inhibitor
(MG) within 6 hr of recovery. MG formation may be an affords neuroprotection with delayed administration in a rat model of
adaptive and reversible response to unfavorable environ- neonatal hypoxic-ischemic brain injury. J Clin Invest 101:1992–1999.
ments at the subcellular level that leads to a decrease in Chung YH, Shin CM, Kim MJ, Lee EY, Kim G, Cha CI. 2002. Enhanced
oxygen consumption and ROS production (Karbowski et expression of p53 in reactive astrocytes following transient focal ischemia.
al., 1999). The third and last step, death-warrant, occurs Neurol Res 24:324 –328.
after 24 hr of reperfusion when a complete disruption of Culmsee C, Bondada S, Mattson MP. 2001a. Hippocampal neurons of mice
free radical-induced MG leads to cyt-c release, caspase deficient in DNA-dependent protein kinase exhibit increased vulnerabil-
activation, and cell death. ity to DNA damage, oxidative stress and excitotoxicity. Brain Res Mol
Brain Res 87:257–262.
It is now accepted generally that massive neuronal Culmsee C, Zhu X, Yu QS, Chan SL, Camandola S, Guo Z, Greig NH,
death due to oxidative stress is a regular feature in neuro- Mattson MP. 2001b. A synthetic inhibitor of p53 protects neurons against
degenerative diseases of the brain. Much less attention, death induced by ischemic and excitotoxic insults, and amyloid beta-
however, has been given to glial cells death. Several studies peptide. J Neurochem 77:220 –228.
suggest that glial cells in neurodegenerative diseases (i.e., de la Monte SM, Sohn YK, Ganju N, Wands JR. 1998. P53- and CD95-
Alzheimer’s disease) are affected more than neurons by associated apoptosis in neurodegenerative diseases. Lab Invest 78:401–
apoptotic cell death (Lassmann et al., 1995; Smale et al., 411.
1995). de la Monte SM, Sohn YK, Wands JR. 1997. Correlates of p53- and Fas
In our glial system, we highlight for the first time (CD95)-mediated apoptosis in Alzheimer’s disease. J Neurol Sci 152:73–
that glial cells respond to oxidative stress undergoing ap- 83.
Ding HF, Fisher DE. 1998. Mechanisms of p53-mediated apoptosis. Crit
optosis via p53 transcription independent pathways, which
Rev Oncog 9:83–98.
is summarized in Figure 9. Based on these results, drugs Ding HF, McGill G, Rowan S, Schmaltz C, Shimamura A, Fisher DE.
such as PF-␣ that selectively protect neurons from apo- 1998. Oncogene-dependent regulation of caspase activation by p53 pro-
ptotic stimuli would not be sufficient in the treatment of tein in a cell-free system. J Biol Chem 273:28378 –28383.
neurodegenerative diseases. Therefore, therapeutic ap- Eskes R, Antonsson B, Osen-Sand A, Montessuit S, Richter C, Sadoul R,
proaches based on caspase inactivation (Cheng et al., 1998; Mazzei G, Nichols A, Martinou JC. 1998. Bax-induced cytochrome C
Robertson et al., 2000, 2001) are likely a more efficient release from mitochondria is independent of the permeability transition
strategy against neurodegenerative diseases because in cell pore but highly dependent on Mg2⫹ ions. J Cell Biol 143:217–224.
death execution, both glial cells and neurons exhibit a final Eskes R, Desagher S, Antonsson B, Martinou JC. 2000. Bid induces the
common pathway in which caspases are the main actors. oligomerization and insertion of Bax into the outer mitochondrial mem-
brane. Mol Cell Biol 20:929 –935.
Gottlieb TM, Oren M. 1998. p53 and apoptosis. Semin Cancer Biol
ACKNOWLEDGMENTS
8:359 –368.
We thank Dr. S. Biocca (University of Rome Tor Green DR. 1998. Apoptotic pathways: the roads to ruin. Cell 94:695– 698.
Vergata) for the gift of C6 cells and for critical discussions. Haupt Y, Barak Y, Oren M. 1996. Cell type-specific inhibition of p53-
This work was supported in part by research fellowships mediated apoptosis by mdm2. EMBO J 15:1596 –1606.
Astrocyte Apoptosis and p53 95

Haupt Y, Maya R, Kazaz A, Oren M. 1997. Mdm2 promotes the rapid gene, p53, is induced in injured brain regions following experimental
degradation of p53. Nature 387:296 –299. traumatic brain injury. Brain Res Mol Brain Res 71:78 – 86.
Jenner P. 2003. Oxidative stress in Parkinson’s disease. Ann Neurol Pochampally R, Fodera B, Chen L, Lu W, Chen J. 1999. Activation of an
53(Suppl):26 –38. MDM2-specific caspase by p53 in the absence of apoptosis. J Biol Chem
Jordan J, Galindo MF, Prehn JH, Weichselbaum RR, Beckett M, Ghadge 274:15271–15277.
GD, Roos RP, Leiden JM, Miller RJ. 1997. p53 expression induces Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. 1997. A model for
apoptosis in hippocampal pyramidal neuron cultures. J Neurosci 17:1397– p53-induced apoptosis. Nature 389:300 –305.
1405. Reed JC, Jurgensmeier JM, Matsuyama S. 1998. Bcl-2 family proteins and
Joseph JA, Strain JG, Jimenez ND, Fisher D. 1997. Oxidant injury in PC12 mitochondria. Biochim Biophys Acta 1366:127–137.
cells—a possible model of calcium “dysregulation” in aging: I. Selectivity Richter-Landsberg C, Vollgraf U. 1998. Mode of cell injury and death after
of protection against oxidative stress. J Neurochem 69:1252–1258. hydrogen peroxide exposure in cultured oligodendroglia cells. Exp Cell
Kannan K, Amariglio N, Rechavi G, Givol D. 2000. Profile of gene Res 244:218 –229.
expression regulated by induced p53: connection to the TGF-beta family. Robertson J, Beaulieu JM, Doroudchi MM, Durham HD, Julien JP,
FEBS Lett 470:77– 82. Mushynski WE. 2001. Apoptotic death of neurons exhibiting peripherin
Karbowski M, Kurono C, Wozniak M, Ostrowski M, Teranishi M, Nish- aggregates is mediated by the proinflammatory cytokine tumor necrosis
izawa Y, Usukura J, Soji T, Wakabayashi T. 1999. Free radical-induced factor-alpha. J Cell Biol 155:217–226.
megamitochondria formation and apoptosis. Free Radic Biol Med 26: Robertson GS, Crocker SJ, Nicholson DW, Schulz JB. 2000. Neuropro-
396 – 409. tection by the inhibition of apoptosis. Brain Pathol 10:283–292.
Kitamura Y, Ota T, Matsuoka Y, Tooyama I, Kimura H, Shimohama S, Sansome C, Zaika A, Marchenko ND, Moll UM. 2001. Hypoxia death
Nomura Y, Gebicke-Haerter PJ, Taniguchi T. 1999. Hydrogen stimulus induces translocation of p53 protein to mitochondria. Detection
peroxide-induced apoptosis mediated by p53 protein in glial cells. Glia by immunofluorescence on whole cells. FEBS Lett 488:110 –115.
25:154 –164. Schuler M, Bossy-Wetzel E, Goldstein JC, Fitzgerald P, Green DR. 2000.
Komarov PG, Komarova EA, Kondratov RV, Christov-Tselkov K, Coon p53 induces apoptosis by caspase activation through mitochondrial cyto-
JS, Chernov MV, Gudkov AV. 1999. A chemical inhibitor of p53 that chrome c release. J Biol Chem 275:7337–7342.
protects mice from the side effects of cancer therapy. Science 285:1733– Schuler M, Green DR. 2001. Mechanisms of p53-dependent apoptosis.
1737. Biochem Soc Trans 29:684 – 688.
Kubbutat MH, Jones SN, Vousden KH. 1997. Regulation of p53 stability Smale G, Nichols NR, Brady DR, Finch CE, Horton WEJ. 1995. Evi-
by Mdm2. Nature 387:299 –303. dence for apoptotic cell death in Alzheimer’s disease. Exp Neurol 133:
Lakkaraju A, Dubinsky JM, Low WC, Rahman YE. 2001. Neurons are 225–230.
protected from excitotoxic death by p53 antisense oligonucleotides de- Soto AM Sonnenschein C. 1985. The role of estrogens on the proliferation
livered in anionic liposomes. J Biol Chem 276:32000 –32007. of human breast tumor cells (MCF-7). J Steroid Biochem 23:87–94.
Lassmann H, Bancher C, Breitschopf H, Wegiel J, Bobinski M, Jellinger K, Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM,
Wisniewski HM. 1995. Cell death in Alzheimer’s disease evaluated by Mangion J, Jacotot E, Costantini P, Loeffler M, Larochette N, Goodlett
DNA fragmentation in situ. Acta Neuropathol (Berl) 89:35– 41. DR, Aebersold R, Siderovski DP, Penninger JM, Kroemer G.
Love S. 2003. Apoptosis and brain ischaemia. Prog Neuropsychopharmacol 1999. Molecular characterization of mitochondrial apoptosis-inducing
Biol Psychiatry 27:267–282. factor. Nature 397:441– 446.
Marchenko ND, Zaika A, Moll UM. 2000. Death signal-induced localiza- Susin SA, Zamzami N, Kroemer G. 1998. Mitochondria as regulators of
tion of p53 protein to mitochondria. A potential role in apoptotic apoptosis: doubt no more. Biochim Biophys Acta 1366:151–165.
signaling. J Biol Chem 275:16202–16212. Tatton WG, Chalmers-Redman R, Brown D, Tatton N. 2003. Apoptosis
Matsuyama S, Reed JC. 2000. Mitochondria-dependent apoptosis and in Parkinson’s disease: signals for neuronal degradation. Ann Neurol
cellular pH regulation. Cell Death Differ 7:1155–1165. 53(Suppl):61–70.
Mercanti D, Luzzatto E, Ciotti MT, Levi G. 1987. Mitogenic effect of a Teranishi M, Spodonik JH, Karbowski M, Kurono C, Soji T, Wakabayashi
human placental factor on astrocytes and glial precursors. Exp Cell Res T. 2000. Swelling of free-radical-induced megamitochondria causes ap-
168:182–190. optosis. Exp Mol Pathol 68:104 –123.
Miyashita T, Reed JC. 1995. Tumor suppressor p53 is a direct transcrip- Volonte C, Ciotti MT, Battistini L. 1994. Development of a method for
tional activator of the human bax gene. Cell 80:293–299. measuring cell number: application to CNS primary neuronal cultures.
Momand J, Wu HH, Dasgupta G. 2000. MDM2—master regulator of the Cytometry 17:274 –276.
p53 tumor suppressor protein. Gene 242:15–29. Wu X, Bayle JH, Olson D, Levine AJ. 1993. The p53-mdm-2 autoregu-
Momand J, Zambetti GP, Olson DC, George D, Levine AJ. 1992. The latory feedback loop. Genes Dev 7:1126 –1132.
mdm-2 oncogene product forms a complex with the p53 protein and Xiang H, Kinoshita Y, Knudson CM, Korsmeyer SJ, Schwartzkroin PA,
inhibits p53-mediated transactivation. Cell 69:1237–1245. Morrison RS. 1998. Bax involvement in p53-mediated neuronal cell
Morrison RS, Kinoshita Y, Johnson MD, Guo W, Garden GA. 2003. death. J Neurosci 18:1363–1373.
p53-dependent cell death signaling in neurons. Neurochem Res 28:15– Zauberman A, Barak Y, Ragimov N, Levy N, Oren M. 1993. Sequence-
27. specific DNA binding by p53: identification of target sites and lack of
Napieralski JA, Raghupathi R, McIntosh TK. 1999. The tumor-suppressor binding to p53–MDM2 complexes. EMBO J 12:2799 –2808.

Potrebbero piacerti anche