Skip to main content
American Journal of Physiology - Lung Cellular and Molecular Physiology logoLink to American Journal of Physiology - Lung Cellular and Molecular Physiology
. 2009 Sep 18;297(5):L945–L953. doi: 10.1152/ajplung.00081.2009

Deletion of caveolin-1 protects hyperoxia-induced apoptosis via survivin-mediated pathways

Meng Zhang 1, Ling Lin 1, Seon-Jin Lee 1, Li Mo 1, Jiaofei Cao 1, Emeka Ifedigbo 1, Yang Jin 1,
PMCID: PMC2777493  PMID: 19767411

Abstract

Hyperoxia-induced lung injury is an established model that mimics human acute respiratory distress syndrome. Cell death is a prominent feature in lungs following prolonged hyperoxia. Caveolae are omega-shaped invaginations of the plasma membrane. Caveolin-1 (cav-1), a 22-kDa transmembrane scaffolding protein, is the principal structural component of caveolae. We have recently shown that deletion of cav-1 (cav-1−/−) protected against hyperoxia-induced cell death and lung injury both in vitro and in vivo; however, the mechanisms remain unclear. Survivin, a member of the inhibitor of apoptosis protein family, inhibits apoptosis in tumor cells. Although emerging evidence suggests that survivin is involved in wound healing, especially in vascular injuries, its role in hyperoxia-induced lung injury has not been investigated. Our current data demonstrated that hyperoxia induced apoptosis via suppressing survivin expression. Deletion of cav-1 abolished this suppression and subsequently protected against hyperoxia-induced apoptosis. Using “gain” and “loss” of function assays, we determined that survivin protected lung cells from hyperoxia-induced apoptosis via the inhibition of apoptosis executor caspase-3. Overexpression of survivin by deletion of cav-1 was regulated by Egr-1. Egr-1 functioned as a negative regulator of survivin expression. Deletion of cav-1 upregulated survivin via decreased Egr-1 binding of the survivin promoter region. Together, this study illustrates the effect of hyperoxia on survivin expression and the role of survivin in hyperoxia-induced apoptosis. We also demonstrate that deletion of cav-1 protects hyperoxia-induced apoptosis via modulation of survivin expression.

Keywords: caspase-3, Egr-1


Acute Lung Injury (ALI) and its more severe manifestation, acute respiratory distress syndrome (ARDS), are devastating conditions that account for high morbidity and mortality among critically ill patients (28, 44). Exposure to high O2 tension (hyperoxia) has been shown in animal models to induce lung injury that closely resembles ARDS. Hyperoxia triggers an extensive inflammatory response in the lung that is typically followed by severe damage of the alveolar-capillary barrier. This damage results in impaired gas exchange and pulmonary edema (10, 30, 47). Cell death of pulmonary capillary endothelial cells and alveolar epithelial cells is the major pathological change in hyperoxia-injured lungs (1, 13, 14, 26). Compromised epithelial and endothelial cell function leads to fluid and macromolecule accumulation in the air space and can cause clinical respiratory failure and death (1, 13, 14, 26, 47). The mechanisms underlying lung injury and cell death in response to hyperoxia remain incompletely understood, although recent studies have implicated that both necrosis and apoptosis pathways play important roles in hyperoxic lung injury (1, 13, 14, 26, 47).

Caveolae are one subset of lipid rafts that are enriched in cholesterol and sphingomyelin and are characterized by their omega-shaped invaginations (50–100 nm) in the plasma membrane (36). These structures participate in a number of cellular processes, including endocytosis, transcytosis, and intracellular signal transduction (2, 25, 36, 39). Caveolin-1 (cav-1), a 22-kDa transmembrane scaffolding protein, serves as the principal structural component of caveolae (2, 25, 36, 39). Recent data indicate that cav-1 mediates ALI via a number of different mechanisms, such as increasing hyperoxic cell death and vascular permeability (16, 20, 22). It is known that deletion of cav-1 protects hyperoxia-induced cell death and ALI both in vitro and in vivo (22); however, the mechanisms remain unclear.

Cell death including apoptosis, necrosis, and oncosis, are features commonly found in ALI and are thought to be one of the mechanisms responsible for ALI/ARDS (10, 28, 30, 44, 47). Apoptosis results from the activation of the family of caspases. Caspases are enzymes that cleave cellular proteins at aspartic acid residues (40). Both the intrinsic and extrinsic apoptotic pathways use caspase-3 as the “executioner” caspase, which cleaves the essential substrates for cell survival (6, 11, 40). Among the antiapoptotic factors, there is a family of proteins named inhibitor of apoptosis proteins (IAP). Survivin, a 16.5-kDa protein, is the smallest member of the IAP family (45). Survivin contains a baculovirus inhibitor of apoptosis repeat protein domain (32, 34, 45) that inhibits apoptosis either directly or indirectly by interfering with the function of caspases (32, 34, 45). Survivin is expressed abundantly in embryonic organs, but only negligible levels can be found in the majority of well-differentiated adult normal tissues (9). Survivin is, however, highly reexpressed in various human cancers (9). Due to its importance in inhibiting cell death and cell division, survivin has been studied extensively as a useful diagnostic marker of cancer and a potential target for cancer treatment (9). Despite the demonstration of a variety of roles attributed to survivin in tumor biology, the function of this protein in normal tissue remains unclear.

Recently, emerging evidence suggests that survivin is involved in tissue injury and wound healing. In vascular injuries, survivin plays multiple roles, including the regulation of vascular cell responses, PDGF gene expression, and vein graft remodeling (5). In neighboring endothelial cells, survivin functions as a critical regulator of angiogenesis (5). In addition, survivin was found to regulate smooth muscle cell growth and may play an important role in the prevention of restenosis following revascularization procedures (19). Although a significant number of studies have explored the role of survivin in cancer research and carcinogenesis, thus far, to our knowledge, no studies have explored the role of survivin in hyperoxia-induced lung injury or have determined whether hyperoxia mediates survivin expression and gene regulation.

Our current study explores the role of deletion of cav-1 in hyperoxic apoptosis and the potential apoptotic pathways involved in lung cells. We report here the discovery of a novel target of hyperoxia in the oxidative apoptosis pathways in lung cells and discuss the potential functions of survivin in cav-1-mediated hyperoxic apoptosis.

MATERIALS AND METHODS

Cell culture and treatments.

Human bronchial epithelial cells (Beas-2B), primary mouse lung fibroblasts, and pulmonary endothelial cells were cultured as described (42) and used for experiments after reaching subconfluent monolayers (usually between culture passages 7 and 17). Primary cells were cultured from the lungs of wild-type C57BL/6 mice as previously described (23). Beas-2B lung epithelial cells were purchased from American Type Culture Collection (Manassas, VA) and cultured in the defined medium, DMEM (Cambrex, East Rutherford, NJ). All cells were grown in humidified incubators containing an atmosphere of 5% CO2 and 95% air at 37°C. Cell cultures were exposed to hyperoxia in modular exposure chambers as described (23), using 95% oxygen with 5% CO2.

Reagents.

Survivin antibodies and siRNA were purchased from Santa Cruz Biotechnology (Santa Cruz, CA) and Applied Biosystems (Foster City, CA). Survivin overexpression clones were obtained from Genecopoeia (Germantown, MD). Caspase activity kits were purchased from Calbiochem (Gibbstown, NJ). All other reagents and chemicals were purchased from Sigma (St. Louis, MO).

Animal exposures.

Wild-type C57BL/6 mice, 8–12 wk, were maintained in laminar flow cages in a pathogen-free facility at the University of Pittsburgh. All procedures were performed in accordance with and approved by the Council on Animal Care at the University of Pittsburgh and the National Research Council's Guide for the Humane Care and Use of Laboratory animals. The C57BL/6 mice were purchased from Jackson Laboratories (Bar Harbor, Maine). The animals were exposed to room air or hyperoxia (95% O2, 5% N2).

Transfections and cell viability assays.

Beas-2B cells were transfected with survivin siRNA and control siRNA using transfection reagent (Santa Cruz, CA). Overexpression vectors were transfected with lipoD293 (Signagen, Gaithersburg, MD). Transiently transfected cells were incubated for an additional 24 h and exposed to hyperoxia. After 48 h, cell viability assays were performed using the CellTiter-Glo Luminescent or CellTiter-Blue Cell Viability Assay according to the manufacturer's protocol (Promega, Madison, WI). Briefly, cells were plated into 96-well plates. After transfection and exposure to hyperoxia, cells were washed twice with cold PBS. One-hundred microliters of PBS was added into each well, followed by 100 μl of CellTiter-Glo Substrate. Cells were incubated at room temperature for at least 10 min; luminescent or fluorescent signal was then measured using an Lmax luminometer or Fluorometer (Molecular Devices, Sunnyvale, CA).

Western blot analysis, immunocytochemistry, and immunoprecipitation.

The following antibodies were used for immunoprecipitation and immunoblotting: monoclonal anti-caspase-3 (BD Transduction Laboratories), mono- and polyclonal anti-survivin (Santa Cruz), and polyclonal anti-caspase-3 (Santa Cruz). Western blot analysis or immunocytochemistry were performed as described previously (23).

EMSA.

Double-stranded probe was made from equal amounts of complementary biotin-labeled oligonucleotides (Integrated DNA Technologies, Coralville, IA). Gel mobility shift assays were performed using the Lightshift chemiluminescent electrophoretic mobility shift assay kit (Pierce, Rockford, IL) according to the manufacturer's instructions. Briefly, 4 μg of nuclear proteins were incubated with 50 fmol of biotin end-labeled double-stranded probe for 20 min at room temperature. Electrophoresis was then carried out on 4% native polyacrylamide gels. The protein-DNA complexes were transferred and crosslinked to a nylon membrane. After incubating membranes in blocking buffer, conjugated/blocking buffer, washing buffer, substrate equilibration buffer, and substrate working solution, the membrane was finally exposed to Biomax MR film (Kodak, Rochester, NY). The sequences used for Egr-1 probe are CGTCAGCTCCCCCGCGATCCAC and GCAGTCGAGGGGGCGCTAGGTG.

Quantitative RT-PCR.

RNA was extracted from cells using the TRIzol method (Invitrogen) and was converted to cDNA using the high-capacity cDNA archive kit (Applied Biosystems). Quantitative RT-PCR was performed as described previously (21), whereas probes and primers for Cyr61 and TaqMan Master Mix for gene expression assays were obtained from Applied Biosystems. Gene expression was analyzed by the ΔΔCt method, with 18s rRNA and βGUS as the endogenous control.

Statistical analysis.

All values were expressed as means ± SD from at least three independent experiments. Differences in measured variables between experimental and control groups were assessed by using the Student's t-test (Statview II statistical package; Abacus Concepts, Berkeley, CA). Statistically significant difference was accepted at P < 0.05.

RESULTS

Deletion of cav-1 protected against hyperoxia-induced apoptosis in vitro.

Our previous studies demonstrated that deletion of cav-1 protected against hyperoxia-induced cell death (22). Previous studies published by others have established that hyperoxia induces cell death via both necrosis and apoptosis (47). In this study, we showed that deletion of cav-1 protected murine fibroblasts from hyperoxia-induced apoptosis ( Fig. 1). In wild-type cells, hyperoxia induced caspase-3 activity in a time-dependent manner; these activities were blunted, however, in cav-1−/− fibroblasts. To verify our observation in epithelial cells, we transfected human bronchial Beas-2B cells with scramble control siRNA and cav-1 siRNA and determined caspase-3 activity following 48 h of hyperoxia. Our results (Fig. 1B) demonstrated that cells transfected with control siRNA exhibited significantly elevated caspase-3 activity, which was not observed in cells transfected with cav-1 siRNA.

Fig. 1.

Fig. 1.

Deletion of caveolin-1 (cav-1) protected hyperoxia-induced apoptosis. Cells were treated with hyperoxia (time course), and caspase-3 activity was measured as described in materials and methods. A: wild-type and cav-1−/− fibroblasts were exposed to hyperoxia. Caspase-3 activity was measured. A time course was determined. Open bars, wild-type cells; filled bars, cav-1−/− cells. B: Beas-2B cells were treated with cav-1 siRNA and then exposed to hyperoxia. After 48 h, caspase-3 activity was measured. Open bars, cells exposed to room air; filled bars, cells exposed to hyperoxia. Three independent assays are represented. *+#P < 0.05.

Deletion of cav-1 augmented survivin expression both in vitro and in vivo.

First, we found that hyperoxia suppressed survivin expression in Beas-2B cells in a time-dependent manner (Fig. 2A). We then explored survivin expression in wild-type and cav-1−/− fibroblasts. Interestingly, we found that deletion of cav-1 (cav-1−/− fibroblasts) increased survivin expression at basal levels and after hyperoxia (Fig. 2B). In contrast to decreased survivin expression in wild-type cells following hyperoxia, we observed that hyperoxia had no effects on survivin expression in cav-1−/− fibroblasts (Fig. 2B). To determine if our observation was not exclusive to fibroblasts, we compared survivin expression with and without hyperoxia in homogenized mouse lung tissue. Again, hyperoxia suppressed survivin in a time-dependent manner in wild-type mice, whereas this effect was significantly reduced in cav-1−/− mice (Fig. 2C).

Fig. 2.

Fig. 2.

Effect of hyperoxia on survivin expression. A: Beas-2B cells were exposed to hyperoxia. At each time point, cell lysates were collected, analyzed by Western blot analysis, and blotted with anti-survivin or β-actin as described in materials and methods. B: mouse lung primary fibroblasts were exposed to hyperoxia. After 24 h, cell lysates were collected, and Western blot analyses were performed using anti-survivin, anti-cav-1, or β-actin. C: C57BL/6 mice were exposed to hyperoxia. At each time point, mice lung tissue was collected and homogenized, and Western blot analyses were performed as above. All blots represent at least 3 repeats.

Survivin protected cells from hyperoxic cell death and apoptosis in vitro.

We next explored whether survivin played a role in hyperoxic cell death and apoptosis by “loss” and “gain” of function assays. Initially, we knocked down survivin by transfection of survivin siRNA. Cell viability was evaluated in cells transfected with survivin siRNA after 48 h of hyperoxia (48 h). As shown in Fig. 1A, knocking down survivin significantly enhanced the hyperoxic cell death in Beas-2B cells (Fig. 3A) and mouse lung fibroblasts (data not shown). We confirmed our initial observation by overexpressing survivin in Beas-2B cells. At 48 h posttransfection, cells overexpressing survivin and cells transfected with control vectors were exposed to hyperoxia as described previously. Cell viability was measured using both CellTiter-Glo and trypan blue assays. Significant resistance to hyperoxic cell death was found in cells overexpressing survivin (Fig. 3B), with increased cell proliferation (data not shown).

Fig. 3.

Fig. 3.

Effect of survivin on hyperoxia-induced cell death and apoptosis. The effect of survivin on hyperoxia-induced cell death was evaluated by 1) loss of function assay using siRNA and 2) gain of function assay using survivin overexpressing vectors. A: Beas-2B human bronchial epithelial cells were treated with control siRNA or survivin siRNA. After hyperoxia (48 h), cell viability was measured using CellTiter-blue assay as described in materials and methods. Inset: Western blot analysis indicates successful knockdown of survivin. B: Beas-2B cells were transfected with control vector and survivin overexpressing vector. After hyperoxia (48 h), cell viability was measured as above. Inset: Western blot analysis indicates successful overexpression of survivin. Open bar, cells exposed to room air; filled bar, cells exposed with hyperoxia. *#P < 0.05. Three similar experiments are represented. The effect of survivin on hyperoxia-induced caspase-3 activation was evaluated by 1) loss of function assay using siRNA and 2) gain of function assay using survivin overexpressing vectors, as described in materials and methods. C: Beas-2B cells were treated with control siRNA and survivin siRNA. After hyperoxia (48 h), caspase-3 activity was measured using a colormetric assay (Calbiochem, Gibbstown, NJ) as described in materials and methods. Inset: Western blot analysis indicates successful knockdown of survivin. D: Beas-2B cells were transfected with control vector and survivin overexpressing vector. After hyperoxia (48 h), caspase-3 activity was measured as above. Inset: Western blot analysis indicates successful overexpression of survivin. Open bars, cells exposed to room air; filled bars, cells exposed with hyperoxia. *#P < 0.05. Three similar experiments are represented. E: the effect of cav-1 deficiency in survivin mediated survival after hyperoxia was evaluated. Open bars, wild-type fibroblasts; filled bars, cav-1−/− cells. Cells were transfected with control and survivin siRNA, respectively, and then exposed to hyperoxia. After 48 h, cells were evaluated for caspase-3 activity. *#P < 0.05. At least 3 repeats are represented.

Survivin protected cells against hyperoxia-induced apoptosis.

To examine the role of survivin in hyperoxia-induced apoptosis, we measured caspase-3 activity. As shown in Fig. 3C, caspase-3 activity was induced 48 h after hyperoxia in Beas-2B cells. Cells transfected with survivin siRNA exhibited higher caspase-3 activity compared with cells transfected with control siRNA. Furthermore, overexpression of survivin in these cells blunted the elevation of caspase-3 activity induced by hyperoxia, shown in Fig. 3D. We further evaluated whether survivin overexpression prevents the release of cytochrome c. First, we overexpressed survivin in Beas-2B cells. After hyperoxia, the release of cytochrome c was examined. Survivin overexpression did not significantly modulate cytochrome c release (data not shown).

We next examined whether the elevated survivin in cav-1−/− cells was responsible for the observed resistance to hyperoxic apoptosis in these cells. As illustrated in Fig. 3E, wild-type and cav-1−/− cells were transfected with control siRNA or survivin siRNA and then exposed to hyperoxia. As expected, hyperoxia induced caspase-3 activity in wild-type cells treated with control siRNA. Transfecting cells with survivin siRNA further augmented this effect. In cav-1−/− cells, hyperoxia-induced apoptosis was reduced, as shown previously. Notably, on day 2 and day 3, the protective effects attributed to an absence of cav-1 were abolished by transfecting survivin siRNA (Fig. 3E).

Hyperoxia regulated interaction between survivin and caspase-3 in cav-1−/− cells.

After determining that survivin protected against hyperoxia-induced apoptosis, we sought to examine the mechanism by which survivin mediated this cytoprotective effect. We found that in cav-1−/− cells, hyperoxia increased the physical binding between caspase-3 and survivin at 4 h after hyperoxia (Fig. 4, A and B). Interestingly, at 12 h, survivin expression was significantly decreased, as shown in Fig. 2A.

Fig. 4.

Fig. 4.

Interaction and colocalization of caspase-3 and survivin, with and without hyperoxia. A: colocalization between survivin and caspase-3 in wild-type and cav-1−/− fibroblasts. Cells were exposed to hyperoxia. After 4 h, cells were fixed in 2% paraformaldehyde followed by immunostaining with anti-caspase-3 (rabbit polyclonal antibodies, Santa Cruz) and anti-survivin (mouse monoclonal antibodies, Santa Cruz). Cells were further stained with Hoechst for nucleus definition. Red, survivin; green, caspase-3; blue, nucleus; yellow, merge. Red arrow, colocalized caspase-3 and survivin. At least 3 identical experiments are represented. B: cell lysates were immunoprecipitated with rabbit polyclonal anti-caspase-3 antibodies and then blotted with mouse monoclonal anti-survivin antibodies. Blot represented 3 independent experiments.

Hyperoxia suppressed survivin expression by downregulation of mRNA.

Next, we sought to determine the mechanisms by which hyperoxia suppressed survivin expression. By Taqman real-time PCR assay, hyperoxia suppressed survivin production via transcriptional regulation, as evidenced by a decrease in survivin mRNA (Fig. 5A) in wild-type fibroblasts. Similar results were found in human epithelial Beas-2B cells (data not shown). Conversely, decreases in mRNA survivin levels were not observed in cav-1−/− cells following hyperoxia (Fig. 5A).

Fig. 5.

Fig. 5.

Effect of hyperoxia on the transcriptional regulation of survivin. A: Beas-2B cells were exposed to room air and hyperoxia. After 24 h, RNA was collected, and reverse transcription was performed as described in materials and methods. Taqman real-time PCR was then performed using the manufacturer's protocol. Relative folds of mRNA transcription were indicated. *+#P < 0.05. At least 3 experiments are represented. B: effect of cav-1 deficiency on Egr-1 binding activities by EMSA, with and without hyperoxia. Wild-type and cav-1/ fibroblasts were exposed to room air or hyperoxia. After 4 h, nucleus extract was determined for protein concentration, and binding activity to Egr-1 consensus sequence was determined as described in materials and methods. At least 3 independent experiments are represented. C: expression and translocation of Egr-1 into cell nucleus after hyperoxia in wild-type and cav-1−/− fibroblasts. Wild-type and cav-1−/− fibroblasts were exposed to hyperoxia. After 4 h, cell nucleus was extracted, and Western blot analysis was used to determine Egr-1 translocation. Blots represent at least 3 independent experiments.

Previously published studies indicate that Egr-1 is a negative regulator in the promoter region of survivin (46). We evaluated whether hyperoxia affected Egr-1 binding activity and whether deletion of cav-1 interfered with this process. First, hyperoxia increased Egr-1 binding activity in a time-dependent manner in Beas-2B cells (data not shown). Next, we further found that deletion of cav-1 (cav-1−/−) decreased Egr-1 binding activity at both the basal level and after hyperoxia (Fig. 5B). In addition, there was a higher Egr-1 nuclear translocation in wild-type cells compared with cav-1−/− cells (Fig. 5C).

Egr-1 is a negative regulator of survivin expression in lung cells.

Given that an absence of cav-1 (cav-1−/−) decreased Egr-1 binding activity at both the basal level and after hyperoxia, and that cav-1−/− cells exhibited elevated survivin expression, we explored the possibility that Egr-1 negatively regulated survivin expression in these cells. As shown in Fig. 6A, using Egr-1 knockout cells (Egr-1−/−), we found elevated survivin expression. We then determined whether hyperoxia affected Egr-1 levels. As shown in Fig. 6B, hyperoxia induced Egr-1 in a time-dependent manner. To confirm if Egr-1 negatively regulated survivin mRNA, we treated Beas-2B cells with scrambled control siRNA or Egr-1 siRNA and then exposed these cells to hyperoxia. After 8 h, survivin mRNA was evaluated by Taqman real-time PCR as described previously. Figure 6C showed that knocking down Egr-1 by siRNA enhanced survivin mRNA transcription after hyperoxia. Given that Egr-1 negatively regulated survivin expression and survivin protected hyperoxia-induced cell death, we were interested in examining the viability of Egr-1−/− cells after hyperoxia. As shown in Fig. 6D, as expected, Egr-1−/− cells resisted hyperoxia-induced cell death compared with wild-type cells. Furthermore, we studied whether an antioxidant prevented the hyperoxia-induced upregulation of Egr-1 in wild-type cells. In Beas-2B cells pretreated with 20 mM N-acetyl-cysteine, hyperoxia-induced Egr-1 expression was significantly reduced compared with untreated cells (Fig. 6E).

Fig. 6.

Fig. 6.

Effect of hyperoxia on Egr-1 expression and effect of Egr-1 deficiency on survivin expression. A: effect of Egr-1 deficiency on survivin expression. Wild-type and Egr-1−/− fibroblasts were exposed to hyperoxia. After 24 h, cell lysates were collected and blotted with anti-survivin antibodies. B: effect of hyperoxia on Egr-1 expression. Beas-2B cells were exposed to hyperoxia. After indicated time of exposure, cell lysates were analyzed by Western blot to evaluate Egr-1 expressions. All blots represent at least 3 independent experiments. C: hyperoxia upregulated Egr-1 mRNA. Beas-2B cells were exposed to room air and hyperoxia. After 24 h, RNA was collected, and reverse transcription was performed as described in materials and methods. Taqman real-time PCR was then performed using the manufacturer's protocol. Relative folds of mRNA transcription were indicated. *P < 0.05. At least 3 experiments are represented. D: effect of Egr-1 deficiency on cell viability after hyperoxia. Wild-type and Egr-1−/− fibroblasts were exposed to hyperoxia. After 48 h, cell viability was evaluated as described in materials and methods. *P < 0.05. Three independent assays are represented. E: effect of N-acetyl-cysteine (NAC) on hyperoxia-induced Egr-1 expression. Beas-2B cells were pretreated with 20 mM NAC, followed by exposure to hyperoxia. After 12 h, cell lysates were analyzed by Western blot to evaluate Egr-1 expressions. Blot represents at least 3 independent experiments.

We next examined the survivin-caspase-3 interaction in Egr-1−/− cells. Colocalization and interaction of survivin-caspase-3 in Egr-1−/− cells were higher than those in wild-type cells (Fig. 7).

Fig. 7.

Fig. 7.

Effect of Egr-1 deficiency (Egr-1−/−) on colocalization and interaction between caspase-3 and survivin with and without hyperoxia. Wild-type and Egr-1−/− cells were exposed to hyperoxia. After 4 h, cells were analyzed by confocal microscopy or co-IP assays as described in materials and methods. A: colocalization of survivin and caspase-3 in the absence and presence of hyperoxia. Red, survivin; green, caspase-3; yellow, merge. At least 3 identical experiments are represented. B: cell lysates were immunoprecipitated with rabbit polyclonal anti-caspase-3 antibodies and then blotted with mouse monoclonal anti-survivin antibodies. Blot represented 3 independent experiments.

DISCUSSION

Cell death including apoptosis, necrosis, and oncosis, are features commonly found in ALI and are thought to be one of the mechanisms responsible for ALI/ARDS (10, 28, 30, 44, 47). Our group has previously shown that deletion of cav-1 protects against ALI. Deletion of cav-1 protected both hyperoxia-induced lung injury in vivo and hyperoxic cell death in vitro (22). The mechanisms of this protective effect remain unexplored. In this study, for the first time, we show that deletion of cav-1 protected against hyperoxic apoptosis by modulating the caspase-dependent pathways via upregulation of survivin. Survivin protected hyperoxia-induced cell death by binding to caspase-3. However, this protective effect is likely to delay rather than prevent cell death after hyperoxia. This is based on our observation that overexpressing survivin failed to reduce the release of cytochrome c after hyperoxia (data not shown).

We examined the mechanisms by which deletion of cav-1 induced survivin expression. The early growth response gene (Egr-1) is activated by hyperoxia (24) and has a binding site in the promoter region of survivin (46). Our results indicated that survivin was negatively regulated by Egr-1 after hyperoxia. Deletion of cav-1 reduced Egr-1 nuclear translocation and binding activity, resulting in upregulation of survivin. This was consistent with previous reports showing that survivin is downregulated following exogenous expression of wild-type Egr-1 (46).

Survivin protected against hyperoxia-induced apoptosis. Deletion of cav-1 protected apoptosis via upregulation of survivin by blocking the nuclear translocation of Egr-1 after hyperoxia and subsequently decreasing Egr-1 binding activity. Egr-1 was a negative regulator of survivin and was induced by hyperoxia in a time-dependent manner (Fig. 5C). Hyperoxia induced both Egr-1 translocation and expression. This can partially explain why hyperoxia decreased survivin expression as early as after 12 h of exposure. Besides transcriptional regulation, regulation of survivin can occur at various levels, including protein degradation and intracellular sequestration (17, 33, 43, 48). Whether cav-1 is involved in these levels of regulation requires further exploration.

Several other questions remain to be clarified. First, besides Egr-1, there are many other transcriptional factors involved in survivin regulation (17, 33, 43, 48), such as signal transducer and activator of transcription 3 (STAT3) family (43), the NF-κB (34), the phosphatidylinositol 3-kinase/Akt pathway (37), and insulin-like growth factor I/mTOR signaling (48). These pathways have been reported to upregulate survivin via regulation of mRNA translation (17, 24, 33, 37, 43, 48). The Ras oncogene family and the antiapoptotic factor Wnt-2 (41) also have been reported to play roles in regulating survivin expression. Furthermore, survivin has been reported to be repressed at the transcriptional level by wild-type p53 (29). Whether hyperoxia and cav-1 have any interactions with these pathways to regulate survivin requires further investigation. In addition, previous studies have shown that the mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK), p38 MAPK, and PI3-kinase pathways contributed to the activation of Egr-1 in response to hyperoxia (4, 7, 38, 49). These pathways are likely to regulate cav-1-mediated apoptosis and downregulation of survivin, given that ERK/PI3-kinase are altered in cav-1−/− (12). Second, does deletion of cav-1 and hyperoxia affect other IAPs, such as XIAP, IAP1, or IAP2? To address this question will help to illustrate whether survivin plays a unique role in hyperoxia-induced apoptosis and further help to determine whether survivin will be a specific target for ALI therapy. Third, cav-1 plays a relatively central role in regulating cell death and survival (35); thus, upregulation of survivin may be one, but not the only, mechanism involved in cav-1-mediated apoptosis after hyperoxia.

While we are focusing on cell survival and death, another aspect is cell proliferation. Previous reports have raised a theory that growth arrest is an important defense against oxidative damage (31). Lung cancer cells arrested in G1 phase had less DNA injury and higher cell survival (31). Interestingly, the synthesis and degradation of survivin has been reported to be modulated in a cell cycle-dependent manner. Survivin transcription is most active in G2-M phase. It was reported to stabilize the microtubules and contribute to bipolar spindle formation (3, 8, 27). Survivin has important roles in mitotic regulation (3), and its synthesis and degradation is tightly controlled by cell cycle (3, 8, 27). Therefore, the major known functions of survivin are the regulation of cell cycle and the inhibition of apoptosis (3). More interestingly, both hyperoxia and cav-1 have been reported to be associated with cell cycle. Cav-1 causes cell cycle arrest at G1/S phase and so does hyperoxia (15, 18). While in G1/S phase, less survivin transcription occurred. Therefore, hyperoxia and cav-1 may control cell cycles via regulating survivin.

Last, our current work is conducted in human bronchial epithelial cells and mouse primary fibroblasts. Whether the same observation will be found in other types of cells remains unexplored. Published data (16, 20, 22) and our previous work had indicated that deletion of cav-1 protects from hyperoxia-induced lung injury (22). Although survivin seems to mediate the protective effect of cav-1−/− in hyperoxia-induced apoptosis in vitro, the importance of survivin in hyperoxia-induced lung injury in vivo remains unclear.

GRANTS

This work was supported by National Heart, Lung, and Blood Institute Grant 5K08-HL-085601-02 (Y. Jin) and an ATS unrestricted research grant (Y. Jin).

ACKNOWLEDGMENTS

We acknowledge the editorial assistance of Dr. Janey Desales Whalen, Office of Research, Health Sciences, University of Pittsburgh.

REFERENCES

  • 1.Barazzone C, Horowitz S, Donati YR, Rodriguez I, Piguet PF. Oxygen toxicity in mouse lung: pathways to cell death. Am J Respir Cell Mol Biol 19: 573–581, 1998 [DOI] [PubMed] [Google Scholar]
  • 2.Belanger MM, Gaudreau M, Roussel E, Couet J. Role of caveolin-1 in etoposide resistance development in A549 lung cancer cells. Cancer Biol Ther 3: 954–959, 2004 [DOI] [PubMed] [Google Scholar]
  • 3.Beltrami E, Plescia J, Wilkinson JC, Duckett CS, Altieri DC. Acute ablation of survivin uncovers p53-dependent mitotic checkpoint functions and control of mitochondrial apoptosis. J Biol Chem 279: 2077–2084, 2004 [DOI] [PubMed] [Google Scholar]
  • 4.Beidelschies MA, Huang H, McMullen MR, Smith MV, Islam AS, Goldberg VM, Chen X, Nagy LE, Greenfield EM. Stimulation of macrophage TNFalpha production by orthopaedic wear particles requires activation of the ERK1/2/Egr-1 and NF-kappaB pathways but is independent of p38 and JNK. J Cell Physiol 217: 652–666, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Blanc-Brude OP, Yu J, Simosa H, Conte MS, Sessa WC, Altieri DC. Inhibitor of apoptosis protein survivin regulates vascular injury. Nat Med 8: 987–994, 2002 [DOI] [PubMed] [Google Scholar]
  • 6.Budinger GR, Tso M, McClintock DS, Dean DA, Sznajder JI, Chandel NS. Hyperoxia-induced apoptosis does not require mitochondrial reactive oxygen species and is regulated by Bcl-2 proteins. J Biol Chem 277: 15654–15660, 2002 [DOI] [PubMed] [Google Scholar]
  • 7.Cabodi S, Morello V, Masi A, Cicchi R, Broggio C, Distefano P, Brunelli E, Silengo L, Pavone F, Arcangeli A, Turco E, Tarone G, Moro L, Defilippi P. Convergence of integrins and EGF receptor signaling via PI3K/Akt/FoxO pathway in early gene Egr-1 expression. J Cell Physiol 218: 294–303, 2009 [DOI] [PubMed] [Google Scholar]
  • 8.Carvalho A, Carmena M, Sambade C, Earnshaw WC, Wheatley SP. Survivin is required for stable checkpoint activation in taxol-treated HeLa cells. J Cell Sci 116: 2987–2998, 2003 [DOI] [PubMed] [Google Scholar]
  • 9.Chiou SK, Jones MK, Tarnawski AS. Survivin - an anti-apoptosis protein: its biological roles and implications for cancer and beyond. Med Sci Monit 9: PI25–PI29, 2003 [PubMed] [Google Scholar]
  • 10.Crapo JD. Morphologic changes in pulmonary oxygen toxicity. Annu Rev Physiol 48: 721–731, 1986 [DOI] [PubMed] [Google Scholar]
  • 11.Drakopanagiotakis F, Xifteri A, Polychronopoulos V, Bouros D. Apoptosis in lung injury and fibrosis. Eur Respir J 32: 1631–1638, 2008 [DOI] [PubMed] [Google Scholar]
  • 12.Engelman JA, Chu C, Lin A, Jo H, Ikezu T, Okamoto T, Kohtz DS, Lisanti MP. Caveolin-mediated regulation of signaling along the p42/44 MAP kinase cascade in vivo. A role of the caveolin-scaffolding domain. FEBS Lett 428: 205–211, 1998 [DOI] [PubMed] [Google Scholar]
  • 13.Fink MP. Role of reactive oxygen and nitrogen species in acute respiratory distress syndrome. Curr Opin Crit Care 8: 6–11, 2002 [DOI] [PubMed] [Google Scholar]
  • 14.Freeman BA, Topolosky MK, Crapo JD. Hyperoxia increases oxygen radical production in rat lung homogenates. Arch Biochem Biophys 216: 477–484, 1982 [DOI] [PubMed] [Google Scholar]
  • 15.Galbiati F, Volonté D, Liu J, Capozza F, Frank PG, Zhu L, Pestell RG, Lisanti MP. Caveolin-1 expression negatively regulates cell cycle progression by inducing G(0)/G(1) arrest via a p53/p21(WAF1/Cip1)-dependent mechanism. Mol Biol Cell 12: 2229–2244, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Garrean S, Gao XP, Brovkovych V, Shimizu J, Zhao YY, Vogel SM, Malik AB. Caveolin-1 regulates NF-kappaB activation and lung inflammatory response to sepsis induced by lipopolysaccharide. J Immunol 177: 4853–4860, 2006 [DOI] [PubMed] [Google Scholar]
  • 17.Gu L, Chiang KY, Zhu N, Findley HW, Zhou M. Contribution of STAT3 to the activation of survivin by GM-CSF in CD34+ cell lines. Exp Hematol 35: 957–966, 2007 [DOI] [PubMed] [Google Scholar]
  • 18.Helt CE, Staversky RJ, Lee YJ, Bambara RA, Keng PC, O'Reilly MA. The Cdk and PCNA domains on p21Cip1 both function to inhibit G1/S progression during hyperoxia. Am J Physiol Lung Cell Mol Physiol 286: L506–L513, 2004 [DOI] [PubMed] [Google Scholar]
  • 19.Hoel AW, Wang GJ, Simosa HF, Conte MS. Regulation of vascular smooth muscle cell growth by survivin. Vascular 15: 344–349, 2007 [DOI] [PubMed] [Google Scholar]
  • 20.Hu G, Ye RD, Dinauer MC, Malik AB, Minshall RD. Neutrophil caveolin-1 expression contributes to mechanism of lung inflammation and injury. Am J Physiol Lung Cell Mol Physiol 294: L178–L186, 2008 [DOI] [PubMed] [Google Scholar]
  • 21.Jin Y, Kim HP, Cao J, Zhang M, Ifedigbo E, Choi AM. Caveolin-1 regulates the secretion and cytoprotection of Cyr61 in hyperoxic cell death. FASEB J 23: 341–350, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jin Y, Kim HP, Chi M, Ifedigbo E, Ryter SW, Choi AM. Deletion of caveolin-1 protects against oxidative lung injury via up-regulation of heme oxygenase-1. Am J Respir Cell Mol Biol 39: 171–179, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Jin Y, Kim HP, Ifedigbo E, Lau LF, Choi AM. Cyr61 protects against hyperoxia-induced cell death via Akt pathway in pulmonary epithelial cells. Am J Respir Cell Mol Biol 33: 297–302, 2005 [DOI] [PubMed] [Google Scholar]
  • 24.Jones N, Agani FH. Hyperoxia induces Egr-1 expression through activation of extracellular signal-regulated kinase 1/2 pathway. J Cell Physiol 196: 326–333, 2003 [DOI] [PubMed] [Google Scholar]
  • 25.Kurzchalia TV, Dupree P, Parton RG, Kellner R, Virta H, Lehnert M, Simons K. VIP21, a 21-kD membrane protein is an integral component of trans-Golgi-network-derived transport vesicles. J Cell Biol 118: 1003–1014, 1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lang JD, McArdle PJ, O'Reilly PJ, Matalon S. Oxidant-antioxidant balance in acute lung injury. Chest 122: 314S–320S, 2002 [DOI] [PubMed] [Google Scholar]
  • 27.Lens SM, Wolthuis RM, Klompmaker R, Kauw J, Agami R, Brummelkamp T, Kops G, Medema RH. Survivin is required for a sustained spindle checkpoint arrest in response to lack of tension. EMBO J 12: 2934–2947, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lium B. Adult respiratory distress syndrome (ARDS). Incidence, clinical findings, pathomorphology and pathogenesis. A review. Nord Vet Med 35: 38–47, 1983 [PubMed] [Google Scholar]
  • 29.Lu CD, Altieri DC, Tanigawa N. Expression of a novel antiapoptosis gene, survivin, correlated with tumor cell apoptosis and p53 accumulation in gastric carcinomas. Cancer Res 58: 1808–1812, 1998 [PubMed] [Google Scholar]
  • 30.Mantell LL, Lee PJ. Signal transduction pathways in hyperoxia-induced lung cell death. Mol Genet Metab 71: 359–370, 2000 [DOI] [PubMed] [Google Scholar]
  • 31.McGrath-Morrow SA, Stahl J. Growth arrest in A549 cells during hyperoxic stress is associated with decreased cyclin B1 and increased p21(Waf1/Cip1/Sdi1) levels. Biochim Biophys Acta 1538: 90–97, 2001 [DOI] [PubMed] [Google Scholar]
  • 32.Mita AC, Mita MM, Nawrocki ST, Giles FJ. Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res 14: 5000–5005, 2008 [DOI] [PubMed] [Google Scholar]
  • 33.Nakano J, Huang CL, Liu D, Ueno M, Sumitomo S, Yokomise H. Survivin gene expression is negatively regulated by the p53 tumor suppressor gene in non-small cell lung cancer. Int J Oncol 27: 1215–1221, 2005 [PubMed] [Google Scholar]
  • 34.O'Driscoll L, Linehan R, Clynes M. Survivin: role in normal cells and in pathological conditions. Curr Cancer Drug Targets 3: 131–152, 2003 [DOI] [PubMed] [Google Scholar]
  • 35.Razani B, Schlegel A, Liu J, Lisanti MP. Caveolin-1, a putative tumour suppressor gene. Biochem Soc Trans 29: 494–499, 2001 [DOI] [PubMed] [Google Scholar]
  • 36.Razani B, Woodman SE, Lisanti MP. Caveolae: from cell biology to animal physiology. Pharmacol Rev 54: 431–467, 2002 [DOI] [PubMed] [Google Scholar]
  • 37.Roca H, Varsos Z, Pienta KJ. CCL2 protects prostate cancer PC3 cells from autophagic death via phosphatidylinositol 3-kinase/AKT-dependent survivin up-regulation. J Biol Chem 283: 25057–25073, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rockel JS, Bernier SM, Leask A. Egr-1 inhibits the expression of extracellular matrix genes in chondrocytes by TNFalpha-induced MEK/ERK signalling. Arthriris Res Ther 11: 8, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, Anderson RG. Caveolin, a protein component of caveolae membrane coats. Cell 68: 673–682, 1992 [DOI] [PubMed] [Google Scholar]
  • 40.Schmitt E, Sané AT, Steyaert A, Cimoli G, Bertrand R. The Bcl-xL and Bax-alpha control points: modulation of apoptosis induced by cancer chemotherapy and relation to TPCK-sensitive protease and caspase activation. Biochem Cell Biol 75: 301–314, 1997 [PubMed] [Google Scholar]
  • 41.Shinjyo T, Kurosawa H, Miyagi J, Ohama K, Masuda M, Nagasaki A, Matsui H, Inaba T, Furukawa Y, Takasu N. Ras-mediated up-regulation of survivin expression in cytokine-dependent murine pro-B lymphocytic cells. Tohoku J Exp Med 216: 25–34, 2008 [DOI] [PubMed] [Google Scholar]
  • 42.Slebos DJ, Ryter SW, van der Toorn M, Liu F, Guo F, Baty CJ, Karlsson JM, Watkins SC, Kim HP, Wang X, Lee JS, Postma DS, Kauffman HF, Choi AM. Mitochondrial localization and function of heme oxygenase-1 in cigarette smoke-induced cell death. Am J Respir Cell Mol Biol 36: 409–417, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 43.Vaira V, Lee CW, Goel HL, Bosari S, Languino LR, Altieri DC. Regulation of survivin expression by IGF-1/mTOR signaling. Oncogene 26: 2678–2684, 2007 [DOI] [PubMed] [Google Scholar]
  • 44.Vasudevan A, Lodha R, Kabra SK. Acute lung injury and acute respiratory distress syndrome. Indian J Pediatr 71: 743–750, 2004 [DOI] [PubMed] [Google Scholar]
  • 45.Verhagen AM, Coulson EJ, Vaux DL. Inhibitor of apoptosis proteins and their relatives: IAPs and other BIRPs. Genome Biol 2: REVIEWS3009, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wagner M, Schmelz K, Dörken B, Tamm I. Transcriptional regulation of human survivin by early growth response (Egr)-1 transcription factor. Int J Cancer 122: 1278–1287, 2008 [DOI] [PubMed] [Google Scholar]
  • 47.Wang X, Ryter SW, Dai C, Tang ZL, Watkins SC, Yin XM, Song R, Choi AM. Necrotic cell death in response to oxidant stress involves the activation of the apoptogenic caspase-8/bid pathway. J Biol Chem 278: 29184–29191, 2003 [DOI] [PubMed] [Google Scholar]
  • 48.Wong LH, Brettingham-Moore KH, Chan L, Quach JM, Anderson MA, Northrop EL, Hannan R, Saffery R, Shaw ML, Williams E, Choo KH. Centromere RNA is a key component for the assembly of nucleoproteins at the nucleolus and centromere. Genome Res 17: 1146–1160, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhang L, Zhang ZG, Liu XS, Hozeska-Solgot A, Chopp M. The PI3K/Akt pathway mediates the neuroprotective effect of atorvastatin in extending thrombolytic therapy after embolic stroke in the rat. Arterioscler Thromb Vasc Biol 27: 2470–2475, 2007 [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Lung Cellular and Molecular Physiology are provided here courtesy of American Physiological Society

RESOURCES