Hyperoxia induces retinal vascular endothelial cell apoptosis through formation of peroxynitrite

Xiaolin Gu, Azza B. El-Remessy, Steven E. Brooks, Mohamed Al-Shabrawey, Nai-Tsi Tsai, and Ruth B. Caldwell

Department of Cellular Biology and Anatomy, Department of Ophthalmology, Department of Pharmacology and Toxicology, and Vascular Biology Center, The Medical College of Georgia, Augusta, Georgia 30912

Submitted 16 September 2002 ; accepted in final form 30 April 2003


    ABSTRACT
 TOP
 ABSTRACT
 MATERIAL AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Hyperoxia exposure induces capillary endothelial cell apoptosis in the developing retina, leading to vaso-obliteration followed by proliferative retinopathy. Previous in vivo studies have shown that endothelial nitric oxide synthase (NOS3) and peroxynitrite are important mediators of the vaso-obliteration. Now we have investigated the relationship between hyperoxia, NOS3, peroxynitrite, and endothelial cell apoptosis by in vitro experiments using bovine retinal endothelial cells (BREC). We found that BREC exposed to 40% oxygen (hyperoxia) for 48 h underwent apoptosis associated with activation of caspase-3 and cleavage of the caspase substrate poly(ADP-ribose) polymerase. Hyperoxia-induced apoptosis was associated with increased formation of nitric oxide, peroxynitrite, and superoxide anion and was blocked by treatment with uric acid, nitro-L-arginine methyl ester, or superoxide dismutase. Analyses of the phosphatidylinositol 3-kinase/Akt kinase survival pathway in cells directly treated with peroxynitrite revealed inhibition of VEGF- and basic FGF-induced activation of Akt kinase. These results suggest that hyperoxia-induced formation of peroxynitrite induces BREC apoptosis by crippling key survival pathways and that blocking peroxynitrite formation prevents apoptosis. These data may have important clinical implications for infants at risk of retinopathy of prematurity.

oxygen-induced retinopathy; vaso-obliteration; superoxide; nitric oxide


OXYGEN TOXICITY is known to play a key role in the pathogenesis of retinopathy of prematurity (ROP), a potentially blinding disorder affecting premature infants. Retinal capillary damage caused by exposing the developing retina to high oxygen tensions is an early initiating event, leading to a poorly controlled process of retinal vascularization and the development of proliferative retinopathy (9). Accordingly, the mechanisms by which oxygen injures developing capillary networks are of major clinical significance.

Alon et al. (2) showed that hyperoxia-induced vasoobliteration occurs via selective apoptosis of retinal vascular endothelial cells. They (2) and other investigators (46) have proposed that premature downregulation of vascular endothelial growth factor (VEGF), an endothelial cell-specific mitogen and survival factor, leads to vaso-obliteration of newly formed capillaries. It also has been shown that hyperoxia causes increased expression of pigment epithelium-derived growth factor (PEDF), a potent angiostatic factor, within the retina (15). A role for direct oxidative damage to endothelial cells by reactive oxygen species (ROS) is supported by studies showing that pretreatment of newborn rats with intravitreal liposome-encapsulated superoxide dismutase (SOD) (41) or systemically administered Trolox (a scavenger of peroxynitrite) (45) diminished the severity of oxygen-induced retinopathy. A role for ROS in ROP is also supported by the fact that antioxidants are in low reserve at early stages of retinal development (42) and that infants with active ROP have low serum levels of reduced glutathione (GSH) (43).

We have shown previously that exposure of neonatal mice to 75% oxygen for 48 h causes degeneration of the retinal capillaries associated with formation of the nitric oxide (NO)-derived oxidant peroxynitrite. Both capillary dropout and protein tyrosine nitration were diminished in mice deficient in endothelial NO synthase (NOS3) or treated with nitro-L-arginine methyl ester (L-NAME), implying that peroxynitrite may be an important mediator of hyperoxia-induced vaso-obliteration (9). Peroxynitrite is a highly reactive oxidant generated by the reaction of NO and superoxide anion. It can be generated directly by NOS, especially when the levels of L-arginine are low (3) or under conditions in which tetrahydbiopterin is oxidized (34). Nucleic acids, lipids, and proteins are all potential targets of peroxynitrite-mediated oxidation reactions (4, 20), and apoptosis is one of the ways in which cells may respond to peroxynitrite-mediated injury (1, 6, 35, 55, 58). It seemed plausible, therefore, that hyperoxia-induced apoptosis of capillary endothelial cells in the developing retina could be mediated in part by the formation of peroxynitrite.

The goal of the present study was to test this hypothesis and to determine the role of peroxynitrite/oxidative stress in endothelial cell apoptosis induced by hyperoxia. The specific aims were to determine whether hyperoxia exposure induces protein tyrosine nitration in cultured endothelial cells, whether this alteration has a role in hyperoxia-mediated endothelial cell death, and whether peroxynitrite exposure alters the intracellular signaling pathway for endothelial cell survival. Our experiments using a tissue culture model system have demonstrated that 40% oxygen induces bovine retinal endothelial cell (BREC) apoptosis as indicated by activation of caspase-3, cleavage of the caspase substrate poly-(ADP-ribose) polymerase (PARP), and increases in numbers of cells with nuclear morphology characteristic of apoptosis. Hyperoxia also increases the formation of NO, superoxide, and peroxynitrite. Pretreatment of BREC with a NOS inhibitor (L-NAME) or with scavengers of superoxide or peroxynitrite blocked the hyperoxia-induced apoptosis. We also have shown that exposing cells to exogenous peroxynitrite blocks growth/survival factor [VEGF, basic fibroblast growth factor (bFGF)]-induced activation of the phosphatidylinositol (PI) 3-kinase/Akt kinase cell survival pathway. Our findings corroborate our in vivo data suggesting a role for NO-derived peroxynitrite as a mediator of hyperoxia-induced apoptosis of capillary endothelial cells in the retina.


    MATERIAL AND METHODS
 TOP
 ABSTRACT
 MATERIAL AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Chemicals and reagents. Peroxynitrite was purchased from Upstate Biotechnology (Lake Placid, NY). Hydroethidine (HE) was from Polysciences (Niles, IL). L-NAME and wortmannin were obtained from Sigma-Aldrich (St. Louis, MO). The caspase-3 fluorometric assay kit was from R&D Systems (Minneapolis, MN). Anti-phospho-Akt, anti-Akt, and anti-PARP antibodies and Akt kinase kit were purchased from Cell Signaling Technology (Beverly, MA). Cell culture reagents were purchased from GIBCO BRL (Grand Island, NY), with the exception of fetal bovine serum (FBS), which was purchased from HyClone Laboratories (Logan, UT) and CS-C complete medium, which was from Cell Systems (Kirkland, WA). Other chemicals and reagents were obtained from Sigma-Aldrich.

Cell culture. Primary cultures of BREC were prepared as described previously (18). Cells from passages 4–8 were used in all experiments. Cells were maintained in M199 supplemented with 10% FBS, 10% CS-C complete medium, 2 mM glutamine, 100 U/ml penicillin, and 100 µg/ml streptomycin at 37°C in a humidified CO2 incubator.

Hyperoxia and normoxia treatment. BREC were grown to 80% confluence and switched to serum-free medium. The next day they were placed in a hyperoxic (40% O2, 5% CO2)or normoxic (21% O2, 5% CO2) environment for 48 h unless otherwise indicated. The 40% O2 level was chosen for hyperoxia exposure on the basis of previous research showing that 40% O2 generates significant oxidative stress without inducing toxicity in retinal epithelial cells and fibroblasts (23). The hyperoxia exposure was performed in a humidified incubator modified by installing the PROOX model 110 oxygen regulator (Reming Bioinstruments, Redfield, NY). The oxygen level was continuously monitored using the PROOX oxygen sensor. The oxygen tension in the culture medium was determined by using a blood gas analyzer (GEM Premier 3000; Instrumentation Laboratory, Lexington, MA) and was found to be 232 ± 11.37 mmHg after 48 h in the 40% O2 environment compared with 164 ± 15.36 mmHg in 21% O2.

Peroxynitrite treatment. Stock concentrations of peroxynitrite were freshly prepared in 0.3 N NaOH. An equal amount of 0.3 N NaOH was used for vehicle control experiments. Decomposed peroxynitrite was used in control experiments and was without effect on any of the parameters analyzed.

Caspase-3 activity. A caspase-3 fluorometric assay kit was used to detect apoptosis. Caspase-3 is an intracellular cysteine protease that exists as a proenzyme and becomes activated during the cascade of events that culminates in apoptosis. BREC in serum-free medium were maintained in 21% O2 (normoxia) or 40% O2 (hyperoxia) for 48 h and then incubated in lysis buffer provided with the kit. Cell lysates were centrifuged at 12,500 g for 10 min. The supernatant was collected, samples equated for protein were reacted with the caspase-3 fluorogenic substrate (DEVD-AFC), and fluorescence was measured with a Cytofluor 4000 spectrophotometer (Framingham, MA) with excitation at 400 nm and emission at 505 nm. The results were expressed as fold increase in caspase-3 activity for the hyperoxia- and normoxia-treated cultures above the level of growing cells maintained for 48 h in media with serum.

Hoechst stain. Apoptotic cells in cultures treated as described above were detected by nuclear staining with Hoechst 33258 (2.5 µg/ml in PBS). Cells were reacted with the stain for 30 min at room temperature, washed twice with PBS, and examined and photographed in a Zeiss Axioskop microscope. At least 15 fields were counted for each treatment. Each field contained over 130 cells. Cells with condensed chromatin or with shrunken, irregular, or fragmented nuclei were considered apoptotic. The number of apoptotic nuclei was calculated relative to the total number of nuclei. Total cell density was also calculated for each treatment condition as an indicator of potential differences in cytotoxicity.

Western blotting analysis. BREC were harvested after various treatments and lysed in modified RIPA buffer (20 mM Tris, pH 7.4, 2.5 mM EDTA, 1% Triton X-100, 1% deoxycholate, 0.1% SDS, 40 mM NaF, 10 mM Na4P2O7, and 1 mM PMSF) (5) for 30 min on ice. Insoluble material was removed by centrifugation at 12,000 g at 4°C for 30 min. For analysis of cleaved PARP, NOS1, NOS2, NOS3, phospho-Akt, or Akt, 50 µg of total protein were boiled in 2x Laemmli sample buffer, separated on a 10–12% SDS-polyacrylamide gel by electrophoresis, transferred to nitrocellulose, and reacted with specific antibody. The primary antibody was detected using a horseradish peroxidase-conjugated goat anti-mouse or anti-rabbit antibody and enhanced chemiluminescence (ECL). Intensity of immunoreactivity was measured using densitometry.

Nitrotyrosine formation. Relative amounts of proteins nitrated on tyrosine were measured using slot-blot techniques as described previously (9, 16). Briefly, cellular protein samples prepared as described above were immobilized onto polyvinylidene difluoride membranes by using a slot blot microfiltration unit (Bio-Rad). A dilution series of peroxynitrite-modified BSA (Cayman Chemical, Ann Harper, MI) was loaded to generate a standard curve, and nitrotyrosine was detected using a monoclonal anti-nitrotyrosine antibody (Cayman Chemical) followed by peroxidase-labeled goat anti-mouse IgG and ECL. Relative levels of nitrotyrosine immunoreactivity were determined by densitometry. Comparison with the standard curve generated from peroxynitrite-modified BSA analysis showed that nitrotyrosine formation in normoxia-treated cells was equivalent to 1.0 µg/ml compared with 1.6 µg/ml nitrosylated BSA in the hyperoxia-treated cells and 4.8 µg/ml nitrosylated BSA in cells treated with 1 mM peroxynitrite.

Immunoprecipitation and Akt kinase assay. BREC plated in 10-cm dishes were treated with or without peroxynitrite, wortmannin, VEGF, or bFGF for the desired time. Cell lysates were prepared as described above for immunoblotting. For immunoprecipitation, 500 µg of protein were incubated with immobilized Akt 1G1 monoclonal antibody-containing beads overnight at 4°C with gentle rocking. The beads were washed twice with lysis buffer and kinase buffer (25 mM Tris, pH 7.5, 5 mM {beta}-glycerolphosphate, 2 mM DTT, 0.1 mM Na3VO4, and 10 mM MgCl2) (31) once. Beads were further incubated in 40 µl of kinase buffer supplemented with 200 µM ATP and 1 µg of glycogen synthase kinase (GSK)-3 fusion protein for 30 min at 30°C. The reaction was terminated with 20 µl of 3x SDS sample buffer. After boiling and centrifuging, the supernatant was loaded on 12% SDS-PAGE gel. Western immunoblotting was carried out on the nitrocellulose membrane with phospho-GSK-3{alpha}/{beta} (Ser21/9) antibody as the primary antibody. Horseradish peroxidase-conjugated goat anti-rabbit antibody and ECL were used to detect the primary antibody. Intensity of immunoreactivity was measured using densitometry.

RNA isolation and relative quantitative RT-PCR analysis. Total RNA was isolated from BREC by the guanidine isothiocyanate method, using the RNeasy kit. A total of 2 µg of RNA was reverse transcribed for 60 min at 37°C with 4 units of Omniscript reverse transcriptase, using random primers. One-half microliter of cDNA was added to each PCR reaction, and 18S was used as endogenous standard in PCR reaction. The optimal ratio of 18S:competimers was determined for accurate quantification in the linear range of each RNA. The nucleotide sequences of the oligonucleotide primers used for RT-PCR were selected to span at least one intron-exon splice boundary to eliminate the possibility of amplification of genomic DNA and were as follows: NOS3, 5'-TGCTGCCCAGGACATCTT-3' and 5'-AAGAAGGTGAGAGCCTGG-3'; and NOS2, 5'-ACATGCAGAATGAGTACCGG-3' and 5'-TCAACATCTCCTGGTGGAAC-3'. PCR was performed at 95°C for 15 min, followed by 26 cycles for NOS3 and 32 cycles for NOS2 at 94°C for 1 min, 58°C (NOS3) or 60°C (NOS2) for 30 s, 72°C for 1 min, and then 72°C for 10 min. PCR products were then separated on a 6% acrylamide gel, stained with SYBRE Green, and quantified by densitometry, using the AlphEase imaging analysis system (Alpha Innotech, San Leandro, CA).

Superoxide measurement. Superoxide production was detected using the oxidative fluorescent dye hydroethidine (HE) (39). HE is freely permeable to cells. In the presence of superoxide, HE is oxidized to ethidium bromide (EtBr), which is trapped by intercalating with the DNA (49), resulting in a fluorescence signal that can be quantified by densitometry. Addition of hydrogen peroxide or other oxidants does not significantly increase EtBr fluorescence (10). BREC cultures were treated with either 21% or 40% O2 for 16 h, switched to HEPES buffer (pH 7.4) for 30 min, and then treated with 2 pM HE. The cultures were maintained in the same 21% or 40% O2 environment throughout treatment. After 20 min, the reaction was stopped by switching to fresh HEPES buffer that had been saturated with either 21% or 40% O2. Images were obtained using a laser scanning confocal microscope equipped with a krypton-argon laser. Normoxia- and hyperoxia-treated BREC were processed and imaged in parallel. Laser settings were identical for acquisition of images from both specimens.

Nitrite measurement. A fluorometric assay with the DAN reagent (2,3-diaminonaphthalene) (40) was used to measure the levels of nitrite in the conditioned medium from BREC treated with either 21% or 40% O2. DAN reacts with nitrite under acidic conditions to form 1-(H)-naphthotriazole, a fluorescent product. Briefly, 250 µl of conditioned medium and standards were incubated with 30 µl of DAN reagent (633 µM in 0.67 N HCl) at room temperature in the dark for 15 min. The reaction was stopped by adding 30 µl of 2 N NaOH, and fluorescence was measured using the Cytofluor 4000 spectrophotometer with excitation at 360 nm and emission at 409 nm. Nitrite concentrations in the different samples were calculated using a standard curve generated from a nitrite dilution series of 10 to 0.156 µM.

Statistical analysis. Each experiment was repeated at least three times. Results are expressed as means ± SE. Statistical significance was determined by one- or two-way ANOVA. P values <0.05 were considered significant.


    RESULTS
 TOP
 ABSTRACT
 MATERIAL AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Hyperoxia induces apoptosis in retinal endothelial cells. The effects of hyperoxia on apoptosis were evaluated by determination of caspase-3 activity, Western blot analysis of cleaved PARP, and morphological study of nuclear changes using the nuclear stain Hoechst 33258. Activation of cysteine proteases, especially caspase-3, is a common feature in the execution of programmed cell death, or apoptosis (54). Increased caspase-3 activity is one of the earliest markers of apoptosis. Caspase-3 cleaves a variety of substrate molecules containing the amino acid motif DEVD, including PARP (11). Proteolytic cleavage of PARP from a 116-kDa polypeptide to an 85-kDa fragment is a sensitive marker at the onset of apoptosis. We found that BREC exposed to hyperoxia displayed significantly increased levels of caspase-3 activity (Fig. 1A). Western blot analysis of cleaved PARP revealed that cells cultured in hyperoxia (40% O2, 48 h) showed significant increases in cleaved PARP compared with cells in normoxic controls (P < 0.01, Fig. 1B). These data were further confirmed by nuclear staining with Hoechst 33258, which showed that the hyperoxia-treated cultures had increased numbers of cells showing signs of nuclear fragmentation, chromatin condensation, and blebbing that is typical of apoptotic cells (Fig. 1C).



View larger version (22K):
[in this window]
[in a new window]
 
Fig. 1. Assessment of hyperoxia-induced apoptosis of bovine retinal endothelial cells (BREC). BREC were maintained for 48 h in normoxia or hyperoxia, and apoptosis was analyzed by a fluorometric assay for caspase-3 activity, Western blotting analysis of cleaved poly(ADP-ribose) polymerase (PARP), and Hoechst 33258 staining of apoptosis. Hyperoxia treatment significantly increased the level of caspase-3 activity (A), the amount of cleaved PARP (B), and the numbers of cells with shrunken, irregular, and fragmented nuclei (C, arrows). Insets: representative images. Values are expressed as means ± SE. N, normoxia; H, hyperoxia. *Statistically significant difference (P < 0.05).

 

Hyperoxia increases formation of peroxynitrite, NO, and superoxide. Peroxynitrite is a short-lived molecule at physiological pH, but it produces stable nitration of protein tyrosine residues. Thus nitrotyrosine immunoreactivity can be used to assay for peroxynitrite formation (22, 25, 50). Slot-blot analysis of proteins from BREC with anti-nitrotyrosine antibody revealed an increase of 60–70% in the amount of nitrotyrosine in hyperoxia-treated cells over that found in normoxic controls (Fig. 2). This finding suggests that hyperoxia causes a significant increase in the formation of peroxynitrite.



View larger version (27K):
[in this window]
[in a new window]
 
Fig. 2. Effect of hyperoxia on peroxynitrite formation in BREC. A: immunoblotting analysis with anti-nitrotyrosine antibody was used to analyze the level of peroxynitrite formed in BREC maintained for 48 h in hyperoxia or normoxia. B: densitometric analysis shows that hyperoxia significantly increased the amount of nitrotyrosine immunoreactivity, indicating an increase in peroxynitrite formation. Values are expressed as means ± SE. *Statistically significant difference (P < 0.05).

 

To further evaluate the effects of hyperoxia on formation of reactive nitrogen species, we used quantitative fluorophotometric assay to measure the accumulation of nitrite in the tissue culture media. Increases in media nitrite accumulation are an indicator of NOS activity in forming NO. Nitrite may also accumulate as a breakdown product of peroxynitrite. Our analyses showed a 1.5-fold increase in nitrite levels in the medium from BREC exposed to 40% O2 for 48 h compared with the medium from normoxic controls (Fig. 3).



View larger version (9K):
[in this window]
[in a new window]
 
Fig. 3. Measurement of nitrite formation in BREC. Conditioned media from BREC maintained 48 h in normoxia or hyperoxia were collected and analyzed for nitrite formation using the fluorometric 2,3-diaminonaphthalene (DAN) reaction. Hyperoxia significantly increased the amount of nitrite formation, indicating that there was more nitric oxide (NO) formed in the hyperoxia-treated cells. Values are expressed as means ± SE. *Statistically significant difference (P < 0.05).

 

To see whether the increases in peroxynitrite and nitrite formation were associated with increases in NOS protein expression, we determined the effects of hyperoxia on the expression of different isoforms of NOS after 48 h. Western blot analysis showed a slight increase in NOS3 protein levels in some of the hyperoxia-exposed cultures, but the group differences were not statistically significant (Fig. 3A). Neither NOS1 nor NOS2 was detected in either normoxia- or hyperoxia-treated cultures.

We next evaluated the effects of hyperoxia on expression of NOS mRNA expression. For these analyses, we studied BREC exposed to hyperoxia for 16 h. We chose this time period because we wanted to avoid potential effects of apoptosis on the results. We also reasoned that if increases in NOS expression contribute to increases in formation of NO and peroxynitrite, increases in mRNA levels should occur relatively early during the hyperoxia exposure. However, this analysis showed only a slight increase in NOS3 mRNA compared with normoxic controls (P < 0.05, Fig. 4). There was no change in immunologic NOS (NOS2) expression. Neuronal NOS (NOS1) was not detected. These results suggest that hyperoxia increases NO formation in BREC due to an action increasing NOS3 activity, with little or no effect on NOS expression.



View larger version (19K):
[in this window]
[in a new window]
 
Fig. 4. Effect of hyperoxia on endothelial (NOS3) and immunologic NO synthase (NOS2) expression in BREC. A: Western blot analysis of NOS3 showed no significant change in NOS3 protein levels. Relative quantitative RT-PCR analysis showed a modest increase in NOS3 (B) but not NOS2 (C) mRNA expression in BREC maintained for 16 h in hyperoxia compared with normoxia. Insets: representative images. Values are expressed as means ± SE. *Statistically significant difference (P < 0.05).

 

Because peroxynitrite is a combination product of nitric oxide and superoxide anion, we determined the formation of superoxide anion after hyperoxia exposure. This analysis was also done in cells exposed to hyperoxia for 16 h because DNA fragmentation during apoptosis can alter the HE staining reaction. BREC exposed to 40% O2 for 16 h showed a 3.4-fold increase (P < 0.05) in EtBr fluorescence compared with normoxic controls. This effect was blocked when cells were preincubated with polyethylene glycol-superoxide dismutase (PEG-SOD; 100 U/ml), demonstrating the specificity of the reaction for superoxide anion (Fig. 5).



View larger version (69K):
[in this window]
[in a new window]
 
Fig. 5. Effect of hyperoxia on superoxide formation in BREC. BREC were maintained for 16 h in normoxia (A) or hyperoxia (B), and superoxide anion was detected by confocal microscopy by using the oxidative fluorescent dye hydroethidine (HE). C: densitometric analysis showed a 3.4-fold increase in ethidium bromide fluorescence in cells exposed to hyperoxia, reflecting an increase in superoxide formation. This increase was inhibited by pretreatment with polyethylene glycol-superoxide dismutase (PEG-SOD; 100 U/ml). Values are expressed as means ± SE. *Statistically significant difference (P < 0.05). Scale bar = 10 x 10 µm.

 

Blocking NOS, peroxynitrite, or superoxide inhibits hyperoxia-induced apoptosis. Treatment of BREC with the NOS inhibitor L-NAME (0.5 µM), the superoxide scavenger PEG-SOD (100 U/ml), or the peroxynitrite scavenger uric acid (1 mM) before (30 min) and throughout exposure to hyperoxia (40% O2, 48 h) completely inhibited the increase in apoptosis (Fig. 6). Uric acid binds peroxynitrite but not NO and inhibits the action of peroxynitrite in causing tyrosine nitration (24, 56). Cell counts confirmed that hyperoxia caused a significant decrease in cell density (from 148 ± 6 cells/field in normoxic cultures to 133 ± 18 cells/field in hyperoxic cultures). This decrease in cell number was completely prevented by the inhibitor treatments. Together, these data strongly suggest that the hyperoxia-induced apoptosis in BREC occurs via a peroxynitrite-mediated mechanism.



View larger version (25K):
[in this window]
[in a new window]
 
Fig. 6. Effects of nitro-L-arginine methyl ester (L-NAME), SOD, and uric acid on hyperoxia-induced apoptosis in BREC. Cultures of BREC were pretreated with the NOS inhibitor L-NAME (0.5 µM; A and D), the superoxide scavenger PEG-SOD (100 U/ml; B and D), or the peroxynitrite scavenger uric acid (1 mM; C and D) for 30 min before and throughout exposure to hyperoxic or normoxic conditions (48 h). Caspase-3 activity assay (A–C) or Hoechst 33258 staining of apoptosis (D) was carried out to detect the relative amount of cells undergoing apoptosis. All 3 inhibitors were able to block the hyperoxia-induced increase in caspase-3 activity and to inhibit apoptosis as shown by Hoechst staining. Values are expressed as means ± SE. *Statistically significant difference (P < 0.05).

 

Peroxynitrite alters the PI 3-kinase/Akt kinase signal transduction pathway for endothelial cell survival. Cell growth and cell death are highly regulated processes required for normal vascular development. Increasing evidence indicates that activation of PI 3-kinase, and its downstream target Akt kinase, are involved in antiapoptotic signaling induced by growth/survival factors (27, 33, 44). Because peroxynitrite can inactivate tyrosine kinases by nitration of key tyrosine residues, we hypothesized that peroxynitrite might cause BREC apoptosis by impairing key survival pathways activated by growth/survival factors. To test this possibility, we evaluated the ability of VEGF or bFGF to activate the PI 3-kinase/Akt kinase pathway in BREC treated with peroxynitrite.

As expected, VEGF and bFGF increased the level of phospho-Akt as well as Akt kinase activity in normoxic BREC (Figs. 7 and 8). These stimulatory effects of VEGF and bFGF were substantially inhibited by pretreatment of the cultures with peroxynitrite. Treatment with peroxynitrite alone increased levels of phospho-Akt and Akt kinase activity, though to a lesser extent than either VEGF or bFGF. Wortmannin, a specific PI 3-kinase inhibitor, effectively blocked Akt kinase activation (Fig. 9). The data in Figs. 7 and 8 suggest that peroxynitrite has the capability of interfering with VEGF- and bFGF-induced activation of the PI 3-kinase/Akt kinase transduction pathway, providing a plausible mechanism whereby peroxynitrite generated during prolonged hyperoxia could lead to BREC apoptosis.



View larger version (35K):
[in this window]
[in a new window]
 
Fig. 7. Effect of peroxynitrite on vascular endothelial growth factor (VEGF)-induced Akt activation. BREC were serum starved and pretreated with peroxynitrite (1 mM) or with the vehicle 0.3 N NaOH overnight. Cells were then treated with VEGF (10 ng/ml) for 15 min. Control cells were treated with an equal volume of PBS. Akt activation was detected using anti-phospho-Akt antibody (A) and Akt kinase assay (B). Results indicate that peroxynitrite inhibited VEGF-induced Akt activation. Each blot is 1 representative of 3 repeated experiments. Equal loading was confirmed by Western blotting with anti-Akt antibody (A, bottom). Results of densitometric analyses are shown below each lane. WB, Western blot; ONOO, peroxynitrite; GSK, glycogen synthase kinase. *P < 0.05 vs. control; ^P < 0.05 vs. VEGF.

 


View larger version (47K):
[in this window]
[in a new window]
 
Fig. 8. Effect of peroxynitrite on basic fibroblast growth factor (bFGF)-induced Akt activation. BREC were serum starved and pretreated with peroxynitrite (1 mM) or with the vehicle 0.3 N NaOH overnight. Cells were then treated with bFGF (10 ng/ml) for 15 min, the peak time for bFGF-induced Akt activation. Control cells were treated with an equal volume of PBS. Akt activation was detected using Akt kinase assay. Results indicate that peroxynitrite was able to inhibit bFGF-induced Akt activation. Each blot is 1 representative of 3 repeated experiments. Equal loading was confirmed by Western blotting with anti-Akt antibody (bottom). Results of densitometric analysis are shown below each lane. *P < 0.05 vs. control; ^P < 0.05 vs. bFGF.

 


View larger version (35K):
[in this window]
[in a new window]
 
Fig. 9. Effect of peroxynitrite and phosphatidylinositol (PI) 3-kinase inhibition on VEGF- and bFGF-induced Akt activation. BREC were serum starved and pretreated with peroxynitrite (1 mM), the PI 3-kinase inhibitor wortmannin (300 µg/ml), or both overnight. Cells were then treated with VEGF or bFGF (10 ng/ml) for 15 min. Control cells were treated with an equal amount of vehicle. Akt activation was detected using Akt kinase assay (top). Equal loading was confirmed by Western blotting with anti-Akt antibody (bottom). Results of densitometric analysis are shown below each lane. *P < 0.05 vs. control; ^P < 0.05 vs. VEGF; 'P < 0.05 vs. bFGF.

 


    DISCUSSION
 TOP
 ABSTRACT
 MATERIAL AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
It is well established that hyperoxia induces apoptosis of retinal vascular endothelial cells during early retinal development, leading to oxygen-induced retinopathy (2). The data presented in this study indicate that apoptosis of retinal vascular endothelial cells exposed to chronic hyperoxia occurs via activation of caspase-3 and PARP cleavage in a process involving peroxynitrite formation. To our knowledge, our study is the first to focus on hyperoxia-induced retinal endothelial cell apoptosis in vitro and on the role of peroxynitrite in this process.

Our studies provide evidence that peroxynitrite formed under hyperoxic conditions may be an important mediator of the apoptosis. Specifically, hyperoxia caused an increase in the formation of NO and superoxide. This in turn caused an increase in endothelial cell peroxynitrite, as indicated by increased levels of nitrotyrosine. Although other reactive nitrogen species can produce nitration of tyrosine residues, nitrotyrosine is considered a likely marker of peroxynitrite under conditions of simultaneous production of NO and superoxide (22, 25, 50). In addition, experiments using pharmacological inhibitors to scavenge superoxide or peroxynitrite or to inhibit the activity of NOS all showed a marked reduction in BREC apoptosis. This finding provides further support for the role of peroxynitrite formation in hyperoxia-induced endothelial cell apoptosis. NOS1 and NOS2 proteins were not detected in BREC, and NOS3 protein levels were not altered by hyperoxia exposure. Thus it is considered likely that the hyperoxia-induced increase in NO formation is the result of an increase in NOS3 activity. Hyperoxia-induced activation of NOS in the absence of increases in NOS protein has been observed in rat cerebral cortex (53).

Other investigators (14) have reported that hyperoxia inhibits the proliferation of retinal vascular endothelial cells but not pericytes. Their experiments showed that the endothelial cells failed to recover their proliferative ability after being returned to the room air environment. Although their study showed no evidence of cytotoxic cell injury, cytotoxicity assays do not detect cell death by apoptosis. Thus it is possible that the inhibition of endothelial cell growth they observed actually involved an increase in apoptosis.

Activation of the caspase family of cysteine proteases has been demonstrated to be a central event in apoptosis (13, 21). Caspase-3 is considered to be one of the major downstream caspases of the apoptotic cascade and has been shown to play a critical role in apoptosis induced by a range of stimuli in many cell types, including endothelial cells exposed to peroxynitrite-modified oxidized low-density lipoprotein (LDL) (32). Our results showed that hyperoxia caused significant increases in activity of caspase-3 and in the amount of cleaved PARP, the caspase-3 substrate, as well as in cells with nuclear morphology characteristic of apoptosis. These data are consistent with the hypothesis that exposure of endothelial cells to chronic, mild hyperoxia results in acceleration of apoptosis through a cascade of signaling events resulting in caspase-3 activation and PARP cleavage.

Cell death via apoptosis occurs as a response to oxidative cellular damage in many cell types (12, 19, 51, 57). Peroxynitrite is known to be a much more potent and reactive oxidant than its constituents, NO and superoxide. Although cultured endothelial cells are normally highly resistant to apoptosis (47, 48), peroxynitrite-oxidized LDL has been shown to induce apoptosis of endothelial cells from the aorta and umbilical vein (32). Peroxynitrite-mediated alterations have also been shown to induce apoptosis of vascular endothelial cells in vivo in stroke-prone hypertensive rats (36). The mechanisms of peroxynitrite-induced apoptosis remain unclear. Many growth factors have been shown to prevent apoptosis and promote cell survival through activation of the PI 3-kinase/Akt kinase pathway. Our data demonstrate that peroxynitrite is capable of blocking activation of the PI 3-kinase/Akt kinase pathway in BREC by VEGF and bFGF. Both of these factors are known to be key growth/survival factors in the retina (7, 26, 28, 46, 52). It is possible, therefore, that the pathological effects of peroxynitrite during the vaso-obliteration phase of oxygen-induced retinopathy could be due in part to interference with VEGF- and/or bFGF-mediated survival functions.

Further studies are needed to determine the molecular mechanisms by which peroxynitrite modifies specific signaling pathways. It is known, however, that peroxynitrite can modify tyrosine-containing proteins, such as protein kinase C (30), Mn-SOD (38), and c-SRC tyrosine kinase (37). This modification can inactivate the tyrosine kinase activity of the molecules, blocking their ability to effectively function in signal transduction (8). Peroxynitrite has also been found to induce tyrosine phosphorylation in some experimental settings, and its effect in activating the PI 3-kinase/Akt pathway is well established (for review, see Ref. 29). Our data showing that peroxynitrite pretreatment blocks the effects of VEGF or bFGF in activating the PI 3-kinase/Akt pathway suggest that peroxynitrite also initiates other signaling processes that interfere with the signaling pathway that mediates cell survival. This concept is further supported by our studies of high-glucose-mediated oxidative stress, which have shown that chronic exposure to high glucose or overnight treatment with 100 µM peroxynitrite causes accelerated apoptosis in BREC (17). This effect is accompanied by increases in activity of apoptosis-associated stress protein p38 mitogen-activated protein kinase, whereas the effects of exogenous VEGF in increasing Akt activation and promoting cell survival are inhibited, indicating that cell survival signaling function is impaired.

In summary, our studies suggest that the NO-derived oxidant peroxynitrite plays a key role in hyperoxia-induced retinal capillary endothelial cell apoptosis, possibly by blocking growth factor-induced activation of the PI 3-kinase/Akt kinase survival pathway. Our studies also suggest that effective blockade of peroxynitrite formation may prevent oxygen-induced endothelial cell apoptosis. These data may have important clinical implications for the management of infants at risk for retinopathy of prematurity.


    DISCLOSURES
 
This research was supported by National Eye Institute Grants R01-EY-04618 and R01-EY-11766 and by an unrestricted award from Research to Prevent Blindness to the Department of Ophthalmology.


    FOOTNOTES
 

Address for reprint requests and other correspondence: R. B. Caldwell, Vascular Biology Center, CB3209A, Medical College of Georgia, Augusta, GA 30912-2500 (E-mail: rcaldwel{at}mail.mcg.edu).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.


    REFERENCES
 TOP
 ABSTRACT
 MATERIAL AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
1. Adrie C, Richter C, Bachelet M, Banzet N, Francois D, Dinh-Xuan AT, Dhainaut JF, Polla BS, and Richard MJ. Contrasting effects of NO and peroxynitrites on HSP70 expression and apoptosis in human monocytes. Am J Physiol Cell Physiol 279: C452–C460, 2000.[Abstract/Free Full Text]

2. Alon T, Hemo I, Itin A, Pe'er J, Stone J, and Keshet E. Vascular endothelial growth factor acts as a survival factor for newly formed retinal vessels and has implications for retinopathy of prematurity. Nat Med 1: 1024–1028, 1995.[ISI][Medline]

3. Arnet UA, McMillan A, Dinerman JL, Ballermann B, and Lowenstein CJ. Regulation of endothelial nitric-oxide synthase during hypoxia. J Biol Chem 271: 15069–15073, 1996.[Abstract/Free Full Text]

4. Arteel GE, Briviba K, and Sies H. Protection against peroxynitrite. FEBS Lett 445: 226–230, 1999.[ISI][Medline]

5. Beckman JS, Beckman TW, Chen J, Marshall PA, and Freeman BA. Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA 87: 1620–1624, 1990.[Abstract]

6. Behar-Cohen FF, Heydolph S, Faure V, Droy-Lefaix MT, Courtois Y, and Goureau O. Peroxynitrite cytotoxicity on bovine retinal pigmented epithelial cells in culture. Biochem Biophys Res Commun 226: 842–849, 1996.[ISI][Medline]

7. Bikfalvi A, Klein S, Pintucci G, and Rifkin DB. Biological roles of fibroblast growth factor-2. Endocr Rev 18: 26–45, 1997.[Abstract/Free Full Text]

8. Brito C, Naviliat M, Tiscornia AC, Vuillier F, Gualco G, Dighiero G, Radi R, and Cayota AM. Peroxynitrite inhibits T lymphocyte activation and proliferation by promoting impairment of tyrosine phosphorylation and peroxynitrite-driven apoptotic death. J Immunol 162: 3356–3366, 1999.[Abstract/Free Full Text]

9. Brooks SE, Gu X, Samuel S, Marcus DM, Bartoli M, Huang PL, and Caldwell RB. Reduced severity of oxygen-induced retinopathy in eNOS-deficient mice. Invest Ophthalmol Vis Sci 42: 222–228, 2001.[Abstract/Free Full Text]

10. Carter WO, Narayanan PK, and Robinson JP. Intracellular hydrogen peroxide and superoxide anion detection in endothelial cells. J Leukoc Biol 55: 253–258, 1994.[Abstract]

11. Casciola-Rosen L, Nicholson DW, Chong T, Rowan KR, Thornberry NA, Miller DK, and Rosen A. Apopain/CPP32 cleaves proteins that are essential for cellular repair: a fundamental principle of apoptotic death. J Exp Med 183: 1957–1964, 1996.[Abstract]

12. Choy JC, Granville DJ, Hunt DW, and McManus BM. Endothelial cell apoptosis: biochemical characteristics and potential implications for atherosclerosis. J Mol Cell Cardiol 33: 1673–1690, 2001.[ISI][Medline]

13. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 326: 1–16, 1997.[ISI][Medline]

14. D'Amore PA and Sweet E. Effects of hyperoxia on microvascular cells in vitro. In Vitro Cell Dev Biol 23: 123–128, 1987.[ISI][Medline]

15. Dawson DW, Volpert OV, Gillis P, Crawford SE, Xu H, Benedict W, and Bouck NP. Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Science 285: 245–248, 1999.[Abstract/Free Full Text]

16. El-Remessy A, Abou-Mohamed G, Caldwell RW, and Caldwell RB. High glucose increases tyrosine nitration and superoxide anion formation in endothelial cells: role of eNOS uncoupling and aldose reductase activation. Invest Ophthalmol Vis Sci 44: 3135–3143, 2003.[Abstract/Free Full Text]

17. El-Remessy A, Bartoli M, Fulton D, Caldwell RW, and Caldwell RB. Oxidative stress inactivates the VEGF survival pathway in endothelial cells exposed to high glucose (Abstract). Circulation 106: 281, 2002.

18. Feng Y, Venema VJ, Venema RC, Tsai N, Behzadian MA, and Caldwell RB. VEGF-induced permeability increase is mediated by caveolae. Invest Ophthalmol Vis Sci 40: 157–167, 1999.[Abstract]

19. Forrest VJ, Kang YH, McClain DE, Robinson DH, and Ramakrishnan N. Oxidative stress-induced apoptosis prevented by Trolox. Free Radic Biol Med 16: 675–684, 1994.[ISI][Medline]

20. Groves JT. Peroxynitrite: reactive, invasive and enigmatic. Curr Opin Chem Biol 3: 226–235, 1999.[ISI][Medline]

21. Grutter MG. Caspases: key players in programmed cell death. Curr Opin Struct Biol 10: 649–655, 2000.[ISI][Medline]

22. Halliwell B, Zhao K, and Whiteman M. Nitric oxide and peroxynitrite. The ugly, the uglier and the not so good: a personal view of recent controversies. Free Radic Res 31: 651–669, 1999.[ISI][Medline]

23. Honda S, Hjelmeland LM, and Handa JT. The use of hyperoxia to induce chronic mild oxidative stress in RPE cells in vitro. Mol Vis 7: 63–70, 2001.[ISI][Medline]

24. Hooper DC, Scott GS, Zborek A, Mikheeva T, Kean RB, Koprowski H, and Spitsin SV. Uric acid, a peroxynitrite scavenger, inhibits CNS inflammation, blood-CNS barrier permeability changes, and tissue damage in a mouse model of multiple sclerosis. FASEB J 14: 691–698, 2000.[Abstract/Free Full Text]

25. Ischiropoulos H, Beers MF, Ohnishi ST, Fisher D, Garner SE, and Thom SR. Nitric oxide production and perivascular nitration in brain after carbon monoxide poisoning in the rat. J Clin Invest 97: 2260–2267, 1996.[Abstract/Free Full Text]

26. Itaya H, Hayashi A, Usui S, Hosohata J, Takahashi T, Fujikado T, and Tano Y. Basic fibroblast growth factor inhibits choriocapillaris atrophy in rabbit. Am J Ophthalmol 132: 94–100, 2001.[ISI][Medline]

27. Kennedy SG, Wagner AJ, Conzen SD, Jordan J, Bellacosa A, Tsichlis PN, and Hay N. The PI 3-kinase/Akt signaling pathway delivers an anti-apoptotic signal. Genes Dev 11: 701–713, 1997.[Abstract]

28. Kinoshita M and Shimokado K. Autocrine FGF-2 is responsible for the cell density-dependent susceptibility to apoptosis of HUVEC: a role of a calpain inhibitor-sensitive mechanism. Arterioscler Thromb Vasc Biol 19: 2323–2329, 1999.[Abstract/Free Full Text]

29. Klotz LO, Schroeder P, and Sies H. Peroxynitrite signaling: receptor tyrosine kinases and activation of stress-responsive pathways. Free Radic Biol Med 33: 737–743, 2002.[ISI][Medline]

30. Knapp LT, Kanterewicz BI, Hayes EL, and Klann E. Peroxynitrite-induced tyrosine nitration and inhibition of protein kinase C. Biochem Biophys Res Commun 286: 764–770, 2001.[ISI][Medline]

31. Kong SK, Yim MB, Stadtman ER, and Chock PB. Peroxynitrite disables the tyrosine phosphorylation regulatory mechanism: lymphocyte-specific tyrosine kinase fails to phosphorylate nitrated cdc2(6–20)NH2 peptide. Proc Natl Acad Sci USA 93: 3377–3382, 1996.[Abstract/Free Full Text]

32. Kotamraju S, Hogg N, Joseph J, Keefer LK, and Kalyanaraman B. Inhibition of oxidized low-density lipoprotein-induced apoptosis in endothelial cells by nitric oxide. Peroxyl radical scavenging as an antiapoptotic mechanism. J Biol Chem 276: 17316–17323, 2001.[Abstract/Free Full Text]

33. Kulik G, Klippel A, and Weber MJ. Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt. Mol Cell Biol 17: 1595–1606, 1997.[Abstract]

34. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, Tarpey M, Fukai T, and Harrison DG. Endothelial regulation of vasomotion in apoE-deficient mice: implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation 103: 1282–1288, 2001.[Abstract/Free Full Text]

35. Lin KT, Xue JY, and Wong PY. Mechanisms of peroxynitrite-induced apoptosis in HL-60 cells. Adv Exp Med Biol 469: 569–575, 1999.[ISI][Medline]

36. Ma XL, Gao F, Nelson AH, Lopez BL, Christopher TA, Yue TL, and Barone FC. Oxidative inactivation of nitric oxide and endothelial dysfunction in stroke-prone spontaneous hypertensive rats. J Pharmacol Exp Ther 298: 879–885, 2001.[Abstract/Free Full Text]

37. MacMillan-Crow LA, Greendorfer JS, Vickers SM, and Thompson JA. Tyrosine nitration of c-SRC tyrosine kinase in human pancreatic ductal adenocarcinoma. Arch Biochem Biophys 377: 350–356, 2000.[ISI][Medline]

38. MacMillan-Crow LA and Thompson JA. Tyrosine modifications and inactivation of active site manganese superoxide dismutase mutant (Y34F) by peroxynitrite. Arch Biochem Biophys 366: 82–88, 1999.[ISI][Medline]

39. Miller FJ Jr, Gutterman DD, Rios CD, Heistad DD, and Davidson BL. Superoxide production in vascular smooth muscle contributes to oxidative stress and impaired relaxation in atherosclerosis. Circ Res 82: 1298–1305, 1998.[Abstract/Free Full Text]

40. Misko TP, Schilling RJ, Salvemini D, Moore WM, and Currie MG. A fluorometric assay for the measurement of nitrite in biological samples. Anal Biochem 214: 11–16, 1993.[ISI][Medline]

41. Niesman MR, Johnson KA, and Penn JS. Therapeutic effect of liposomal superoxide dismutase in an animal model of retinopathy of prematurity. Neurochem Res 22: 597–605, 1997.[ISI][Medline]

42. Oguni M, Tanaka O, Tamura H, Shinohara H, Kato K, and Setogawa T. Ontogeny of copper-zinc and manganese superoxide dismutase in the developing rat retina: immunohistochemical and immunochemical study. Ophthalmic Res 27: 227–233, 1995.[ISI][Medline]

43. Papp A, Nemeth I, Karg E, and Papp E. Glutathione status in retinopathy of prematurity. Free Radic Biol Med 27: 738–743, 1999.[ISI][Medline]

44. Parrizas M, Saltiel AR, and LeRoith D. Insulin-like growth factor 1 inhibits apoptosis using the phosphatidylinositol 3'-kinase and mitogen-activated protein kinase pathways. J Biol Chem 272: 154–161, 1997.[Abstract/Free Full Text]

45. Penn JS, Tolman BL, and Bullard LE. Effect of a water-soluble vitamin E analog, trolox C, on retinal vascular development in an animal model of retinopathy of prematurity. Free Radic Biol Med 22: 977–984, 1997.[ISI][Medline]

46. Pierce EA, Foley ED, and Smith LE. Regulation of vascular endothelial growth factor by oxygen in a model of retinopathy of prematurity. Arch Ophthalmol 114: 1219–1228, 1996.[Abstract]

47. Pober JS and Cotran RS. Cytokines and endothelial cell biology. Physiol Rev 70: 427–451, 1990.[Free Full Text]

48. Pohlman TH and Harlan JM. Human endothelial cell response to lipopolysaccharide, interleukin-1, and tumor necrosis factor is regulated by protein synthesis. Cell Immunol 119: 41–52, 1989.[ISI][Medline]

49. Rothe G and Valet G. Flow cytometric analysis of respiratory burst activity in phagocytes with hydroethidine and 2',7'-dichlorofluorescin. J Leukoc Biol 47: 440–448, 1990.[Abstract]

50. Sawa T, Akaike T, and Maeda H. Tyrosine nitration by peroxynitrite formed from nitric oxide and superoxide generated by xanthine oxidase. J Biol Chem 275: 32467–32474, 2000.[Abstract/Free Full Text]

51. Slater AF, Nobel CS, and Orrenius S. The role of intracellular oxidants in apoptosis. Biochim Biophys Acta 1271: 59–62, 1995.[ISI][Medline]

52. Stone J, Itin A, Alon T, Pe'er J, Gnessin H, Chan-Ling T, and Keshet E. Development of retinal vasculature is mediated by hypoxia-induced vascular endothelial growth factor (VEGF) expression by neuroglia. J Neurosci 15: 4738–4747, 1995.[Abstract]

53. Thom SR, Bhopale V, Fisher D, Manevich Y, Huang PL, and Buerk DG. Stimulation of nitric oxide synthase in cerebral cortex due to elevated partial pressures of oxygen: an oxidative stress response. J Neurobiol 51: 85–100, 2002.[ISI][Medline]

54. Thornberry NA and Lazebnik Y. Caspases: enemies within. Science 281: 1312–1316, 1998.[Abstract/Free Full Text]

55. Virag L, Scott GS, Cuzzocrea S, Marmer D, Salzman AL, and Szabo C. Peroxynitrite-induced thymocyte apoptosis: the role of caspases and poly (ADP-ribose) synthetase (PARS) activation. Immunology 94: 345–355, 1998.[ISI][Medline]

56. Whiteman M and Halliwell B. Protection against peroxynitrite-dependent tyrosine nitration and alpha 1-antiproteinase inactivation by ascorbic acid. A comparison with other biological antioxidants. Free Radic Res 25: 275–283, 1996.[ISI][Medline]

57. Yokoyama I, Negita M, Liu DG, Nagasaka T, Kobayashi T, Hayakawa A, Hayashi S, and Nakao A. Prevention of free-radical induced apoptosis by induction of human recombinant Cu, Zn-SOD in pig endothelial cells. Transpl Int 15: 220–225, 2002.[ISI][Medline]

58. Yue G, Lai PS, Yin K, Sun FF, Nagele RG, Liu X, Linask KK, Wang C, Lin KT, and Wong PY. Colon epithelial cell death in 2,4,6-trinitrobenzenesulfonic acid-induced colitis is associated with increased inducible nitric-oxide synthase expression and peroxynitrite production. J Pharmacol Exp Ther 297: 915–925, 2001.[Abstract/Free Full Text]