Reactive oxygen species and caspase activation mediate silica-induced apoptosis in alveolar macrophages

Han-Ming Shen1, Zhuo Zhang2, Qi-Feng Zhang2, and Choon-Nam Ong1

1 Department of Community, Occupational, and Family Medicine, Faculty of Medicine, National University of Singapore, Singapore 117597, Republic of Singapore; and 2 Pneumoconiosis Division, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310013, Peoples Republic of China


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Alveolar macrophages (AMs) are the principal target cells of silica and occupy a key position in the pathogenesis of silica-related diseases. Silica has been found to induce apoptosis in AMs, whereas its underlying mechanisms involving the initiation and execution of apoptosis are largely unknown. The main objective of the present study was to examine the form of cell death caused by silica and the mechanisms involved. Silica-induced apoptosis in AMs was evaluated by terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay and cell cycle/DNA content analysis. The elevated level of reactive oxygen species (ROS), caspase-9 and caspase-3 activation, and poly(ADP-ribose) polymerase (PARP) cleavage in silica-treated AMs were also determined. The results showed that there was a temporal pattern of apoptotic events in silica-treated AMs, starting with ROS formation and followed by caspase-9 and caspase-3 activation, PARP cleavage, and DNA fragmentation. Silica-induced apoptosis was significantly attenuated by a caspase-3 inhibitor, N-acetyl-Asp-Glu-Val-Asp aldehyde, and ebselen, a potent antioxidant. These findings suggest that apoptosis is an important form of cell death caused by silica exposure in which the elevated ROS level that results from silica exposure may act as an initiator, leading to caspase activation and PARP cleavage to execute the apoptotic process.

terminal deoxynucleotidyltransferase-mediated deoxyuridine 5'-triphosphate nick end-labeling assay; poly(adenosine 5'-diphosphate-ribose) polymerase; ebselen; antioxidants


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

CRYSTALLINE SILICA is among the most common and potent occupational fibrogenic agents capable of inducing lung fibrosis and many other lung diseases including lung cancer (11, 33). Although the exact mechanisms involved in the development of pulmonary silicosis and silica-related carcinogenesis have not been fully elucidated, it is generally believed that 1) alveolar macrophages (AMs) are the principal target cells of silica and that they play a critical role in the fibrogenic process in silicosis (11, 26) and 2) reactive oxygen species (ROS) and oxidative damage are closely involved in silica-induced pulmonary damage (33, 39). The importance of AMs is primarily due to their unique role in mediating the interactions between inhaled particulates and various cell types such as lymphocytes and fibroblasts through the release of a wide variety of inflammatory and growth-mediating factors as well as of cytokines (17).

Apoptosis is a distinct form of cell death characterized by cell shrinkage, plasma membrane blebbing, nuclear chromatin condensation, and DNA fragmentation. Different from necrosis, apoptosis is a genetically controlled active cell death process implicated as a critical physiological or pathological mechanism in development and tissue homeostasis as well as in many diseases including cancer (40). Although the details of the mechanisms of apoptosis have not been fully established, a group of cysteine proteases have been identified in both Caenorhabditis elegans (ced-3) and mammalian cells (caspases) as playing a critical role in the apoptotic process (5, 9, 37). Among the various caspases, caspase-9 and caspase-3 appear to be particularly important (22, 29). First, procaspase-9 forms an essential part of the "apoptosome," resulting in caspase-9 activation (14, 24). The activated caspase-9 cleaves downstream caspases such as caspase-3 to orchestrate the biochemical execution of apoptosis. Second, caspase-3 is one of the major effector caspases and plays a critical role in the characteristic apoptotic changes including chromatin condensation, DNA fragmentation, and formation of apoptotic bodies (29).

Although some preliminary studies (19, 20, 23, 30) have shown that silica is able to induce apoptosis in AMs, relatively little is known about the underlying mechanisms involved. Based on the facts that silica induces ROS formation and oxidative stress in AMs and that oxidative stress is an important mediator of apoptosis, it is therefore of interest to investigate the role of ROS and oxidative stress in mediating silica-induced apoptosis in cultured rat AMs. Data from this report demonstrate that silica-induced oxidative stress initiates caspase activation and then triggers the execution of apoptosis.


    EXPERIMENTAL PROCEDURES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Chemicals. Standard crystalline silica was obtained from the Institute of Occupational Medicine, Chinese Academy of Preventive Medicine (Beijing, China) and has been well characterized as being endotoxin free. More than 95% of the particulates were <5 µm in diameter. Lucigenin was purchased from Molecular Probes (Eugene, OR). RPMI 1640 medium and fetal bovine serum were from GIBCO BRL (Life Technologies, Gaithersburg, MD). Ebselen was kindly provided by Rhone-Poulenc Rorer. The caspase-9 substrate N-acetyl-Leu-Glu-His-Asp 7-amino-4-trifluoromethyl coumarin (Ac-LEHD-AFC), 7.5% Ready Gel, and the protein quantification assay kit were from Bio-Rad Laboratories (Hercules, CA). The caspase-3 substrate N-acetyl-Asp-Glu-Val-Asp 7-amino-4-methylcoumarin (Ac-DEVD-AMC) and caspase-3 inhibitor N-acetyl-Asp-Glu-Val-Asp aldehyde (Ac-DEVD-CHO) were from PharMingen (San Diego, CA). Anti-poly(ADP-ribose) polymerase (PARP) was from BIOMOL (Plymouth Meeting, PA). Secondary anti-mouse IgG (peroxidase conjugated) and chemiluminescent substrate were purchased from Pierce (Rockford, IL). Other common chemicals and reagents were purchased from Sigma (St. Louis, MO) and were of analytic or HPLC grade.

Cell culture and treatment. Male Sprague-Dawley rats (body weight 200-220 g) were provided by the Animal Center, National University of Singapore. AMs were collected by lung lavage as previously described (42). After the lavage, the AMs were washed with PBS twice (800 g for 5 min at 37°C) and cultured in RPMI 1640 medium containing 10% fetal bovine serum, 100 U/ml of penicillin, and 100 µg/ml of streptomycin in a 5% CO2 incubator at 37°C.

The stock suspension of silica was prepared in PBS, and the final concentration was in micrograms per 106 cells. Stock ebselen (50 mM) was prepared in DMSO, and it was administered simultaneously with silica at a final concentration of 25 µM. The caspase-3 inhibitor Ac-DEVD-CHO (50 µM) was pretreated with AMs for 1 h before silica exposure.

Lucigenin-dependent chemiluminescence test. Lucigenin-dependent chemiluminescence (CL) measures the formation of superoxide anion radical (O2-·) in AMs. The test was carried out according to an established protocol (32) in our laboratory. The stock solution of 10 mM lucigenin was prepared in PBS and stored at -20°C in the dark. The basic reaction mixture contained 1 × 106 cells and 100 µM lucigenin in 1 ml of PBS. The CL reaction was initiated by the addition of silica, and the CL level was monitored as relative light units (RLU) in a luminometer for a total period of 10 min.

2',7'-Dichlorohydroxyfluorescein diacetate fluorescence test. The level of hydrogen peroxide (H2O2) in AMs was determined with the 2',7'-dichlorohydroxyfluorescein diacetate (DCFH-DA) fluorescence test based on the method described earlier (31). Stock DCFH-DA (5 mM) was dissolved in ethanol and kept at -70°C in the dark. AMs were incubated in 24-well plates, each well containing 1.5 × 105 AMs and 2 µM DCFH-DA in 2 ml of medium. The reaction was initiated by the addition of silica particles and incubated at 37°C for up to 4 h. The fluorescence intensity was measured with a plate reader (Tecan Spectrafluo Plus) with an excitation wavelength of 485 nm and an emission wavelength of 535 nm.

Determination of caspase-9 and caspase-3 activities in silica-treated AMs. Caspase-9 and caspase-3 activities were determined with their cell-permeable synthetic substrates Ac-LEHD-AFC and Ac-DEVD-AMC, respectively. After the designated treatments, AMs were collected from culture flasks and washed twice with PBS before they were lysed in a cell lysis buffer [10 mM Tris · HCl (pH 7.5), 10 mM NaH2PO4 (pH 7.5), 130 mM NaCl, 1% Triton X-100, and 10 mM sodium pyrophosphate]. Both substrates were reconstituted in sterile double-distilled H2O (1 µg/µl) and added to the cell lysate (10 µg/1 ml). The reaction mixture was incubated at 37°C for 1 h in the dark. The fluorescence intensity of AFC or AMC liberated from the substrate was measured with spectrofluorometry (excitation at 390 nm and emission at 510 nm for AFC; excitation at 380 nm and emission at 440 nm for AMC; Perkin-Elmer LS-5B).

PARP cleavage. PARP cleavage was detected by Western blot. After treatment, the cells were collected and lysed in cell lysis buffer containing 150 mM NaCl, 50 mM Tris · HCl, 1% Triton X-100, 1% SDS, 1 mM phenylmethylsulfonyl fluoride, 1 µg/ml of aprotinin, and 1 µg/ml of leupeptin. After denaturation, the samples (30 µg/well) were run on a 7.5% Ready Gel (Bio-Rad) and transferred to a nitrocellulose membrane. Immunoblotting was performed with incubation of the mouse monoclonal anti-PARP (1:5,000) primary antibody followed by incubation with anti-mouse IgG (peroxidase conjugated, 1:5,000). Signal detection was carried out with an enhanced chemiluminescence system and visualized by autoradiography.

Terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay. DNA fragmentation in apoptosis was determined by terminal deoxynucleotidyltransferase (TdT)-mediated dUTP nick end-labeling (TUNEL) assay. The experiment was conducted as reported earlier (32) with minor modifications. After treatment, AMs were collected, washed, and resuspended in PBS with 1% BSA before being fixed with 2% paraformaldehyde for 30 min at room temperature. The fixed cells were then permeabilized with 0.1% Triton X-100 and 0.1% sodium citrate for 3 min on ice. The TUNEL reaction took place with addition of the reaction mixture (containing nucleotides and TdT enzyme) and was incubated for 60 min at 37°C in dark. After one wash with PBS, the cells were finally resuspended in PBS for flow cytometry analysis (Coulter Epics Elite ESP, Miami, FL). At least 10,000 cells were collected in each group. In addition, the changes in cell size (forward scatter) and cell granularity (side scatter) of AMs were also analyzed and recorded. The signal was discriminated against silica particulates and cell debris based on the size. A negative control without the addition of TdT enzymes was always included in each experiment. The data obtained from flow cytometry were analyzed with WinMDI 2.7 software.

Measurement of cell cycle/DNA content. It is well established that DNA fragmentation during apoptosis may lead to extensive loss of DNA content and result in a distinctive sub-G1 peak when analyzed with flow cytometry (27). The cells were first fixed and permeabilized with 70% ice-cold ethanol for >2 h followed by incubation with the freshly prepared staining buffer (0.1% Triton X-100 in PBS, 200 µg/ml of RNase A, and 20 µg/ml of propidium iodide) for 15 min at 37°C. Cell cycle was analyzed with flow cytometry with at least 10,000 cells for each sample. The histogram was abstracted, and the percentage of cells in the sub-G1 phase was then calculated with WinMDI 2.7 software.

Statistical analysis. All data were based on at least three independent experiments; the numerical data are means ± SD and were analyzed with one-way ANOVA with Scheffé's test. A P value < 0.05 was considered significant.


    RESULTS
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Silica-induced ROS formation in AMs. The results in Fig. 1A show that silica-induced O2-· formation was both time and dose dependent. A significant difference between the silica-treated and control groups was observed from the first minute onward. At the end of incubation (10 min), the RLU in AMs treated with the highest concentration of silica (100 µg/106 cells) was more than five times higher than that in the control cells (12,520 vs. 2,240 RLU). Meanwhile, O2-· production in the control cells was maintained at a consistent low level throughout the period. Figure 1B shows the time- and dose-dependent changes in H2O2 formation in silica-treated AMs as determined by the DCFH-DA test. Compared with that in the untreated control cells, all three doses of silica significantly enhanced the dichlorofluorescein (DCF) fluorescence intensity. For instance, at the end of the incubation (4 h), the DCF fluorescence intensity in AMs exposed to a high concentration of silica (100 µg/106 cells) was about two times higher than that in the control cells.


View larger version (17K):
[in this window]
[in a new window]
 
Fig. 1.   Silica-induced reactive oxygen species (ROS) formation in rat alveolar macrophages (AMs) as determined by lucigenin-dependent chemiluminescence (CL) for the detection of O2-· (A) and 2',7'-dichlorohydroxyfluorescein diacetate (DCFH-DA) fluorescence test for the detection of H2O2 (B). Data are means ± SD and were analyzed with 1-way ANOVA with Scheffé's test; n = 4 experiments. Significant difference (P < 0.05) from: # control at all time points; * control at respective time points.

Silica-induced caspase-9 activation in AMs. The enhanced caspase-9 activity in silica-treated AMs was determined by the increase in AFC fluorescence liberated from its substrate Ac-LEHD-AFC. In the time-course study (Fig. 2A), caspase-9 activation was found to be an early event because it started to increase as early as 0.5 h after silica exposure. Caspase-9 activity reached a plateau from 2 h onward. In the dose-response study (Fig. 2B), elevated AFC fluorescence intensity was detected even with the lowest concentration of silica (25 µg/106 cells for 4 h) and increased in a dose-dependent manner.


View larger version (13K):
[in this window]
[in a new window]
 
Fig. 2.   Silica-induced caspase-9 activation in rat AMs as measured by 7-amino-4-trifluoromethyl coumarin (AFC) fluorescence intensity liberated from its substrate N-acetyl-Leu-Glu-His-Asp-AFC (Ac-LEHD-AFC). A: time course. B: dose response. Data are means ± SD; n = 3 experiments. * Significant difference from control group, P < 0.05 by 1-way ANOVA with Scheffé's test.

Silica-induced caspase-3 activation in AMs. The time- and dose-dependent changes in caspase-3 activation in silica-treated AMs were studied, and the results are presented in Fig. 3, A and B, respectively. In the time-course study, caspase-3 activation was found to be an early event because it started to increase as early as 0.5 h after silica exposure. Caspase-3 activity peaked at 2 h and tended to decrease thereafter. In the dose-response study, a significant increase in AMC fluorescence intensity resulting from the cleavage of the caspase-3 substrate Ac-DEVD-AMC was detected even with the lowest concentration of silica (25 µg/106 cells for 4 h). Therefore, caspase-3 showed a trend generally consistent with that of caspase-9.


View larger version (15K):
[in this window]
[in a new window]
 
Fig. 3.   Silica-induced caspase-3 activation in rat AMs as measured by 7-amino-4-methylcoumarin (AMC) fluorescence intensity cleaved from its substrate N-acetyl-Asp-Glu-Val-Asp-AMC (Ac-DEVD-AMC). A: time course. B: dose response. Data are means ± SD; n = 4 experiments. * Significant difference from control group, P < 0.05 by 1-way ANOVA with Scheffé's test.

Silica-induced PARP cleavage in AMs. In the present study, silica-induced PARP cleavage in AMs was determined with Western blotting, and the results are presented in Fig. 4. Evident PARP cleavage (appearance of the 85-kDa band) was observed when cells were treated with silica (100 µg/106 cells) for >1 h or when cells were exposed to silica at concentrations over 50 µg/106 cells for 4 h.


View larger version (23K):
[in this window]
[in a new window]
 
Fig. 4.   Silica-induced poly(ADP-ribose) polymerase (PARP) cleavage in rat AMs as determined by Western blotting. The 116-kDa band is the uncleaved full-length PARP, whereas the 85-kDa band represents the apoptotic cleavage. Equal amount of protein (30 µg) was loaded for each well and subjected to 7.5% Ready Gel.

Silica-induced apoptosis in AMs. In this study, silica-induced apoptosis was evaluated based on the following three measurements: 1) TUNEL assay, 2) cell-cycle/DNA content analysis, and 3) changes in cell size and cell granularity. Figure 5A presents the histograms of both control and silica-treated AMs in the TUNEL assay, showing the increase in TUNEL-positive cells in silica-treated AMs. The time- and dose-dependent increases in TUNEL-positive cells resulting from silica exposure are presented in Fig. 5, B and C, respectively. When treated with a high concentration of silica (100 µg/106 cells), a significant increase in the number of apoptotic cells was observed from 1 h onward and >20% of the cells were apoptotic when treated for 4 h. When the silica concentration increased up to 200 µg/106 cells, ~40% cells were found to be TUNEL positive (data not shown). Moreover, silica-induced apoptosis was further assessed by the determination of cell cycle/DNA content analysis. As shown in Fig. 6A, a distinctive peak of sub-G1 cells was observed in silica-treated cells, whereas no such peak existed in the control group (inset). The percentage of sub-G1 cells was also found to be time and dose dependent as shown in Fig. 6, B and C, respectively, similar to the patterns observed in the TUNEL assay. Last, the results shown in Fig. 7 demonstrate the reduced cell size (forward scatter) and increased cell granularity (side scatter), other characteristic changes of apoptosis, in silica-treated AMs.


View larger version (14K):
[in this window]
[in a new window]
 
Fig. 5.   Silica-induced apoptosis in rat AMs as measured by terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling (TUNEL) assay. A: histograms of the TUNEL results showing the increase in TUNEL-positive cells in silica-treated AMs. B: time course. C: dose response. Data are means ± SD and were analyzed with 1-way ANOVA with Scheffé's test; n = 3 experiments. * Significant difference from control group, P < 0.05.



View larger version (13K):
[in this window]
[in a new window]
 
Fig. 6.   Silica-induced apoptosis in rat AMs as measured by cell cycle/DNA content analysis. A: histograms showing the increase in the distinctive sub-G1 cells in silica-treated AMs compared with that in the control group (inset). PMT4 represents DNA content. B: time course. C: dose response. Data are means ± SD and were analyzed with 1-way ANOVA with Scheffé's test; n = 3 experiments. * Significant difference from control group, P < 0.05.



View larger version (30K):
[in this window]
[in a new window]
 
Fig. 7.   Dot plots showing the changes in cell size (forward scatter) and cell granularity (side scatter) in control (A) and silica-treated (B) AMs. Data were from the TUNEL assay analyzed with flow cytometry. Decreased cell size and increased cell granularity were observed in silica-treated cells.

Effects of Ac-DEVD-CHO on silica-induced caspase-3 activation and apoptosis in AMs. Figure 8A shows that pretreatment of the cells with Ac-DEVD-CHO (50 µM for 2 h) before silica exposure almost completely suppressed caspase-3 activation because the caspase-3 activity in that group was nearly the same as in the control cells. Moreover, Ac-DEVD-CHO also markedly inhibited silica-induced apoptosis in AMs as determined by TUNEL assay (Fig. 8B), thus indicating the involvement of caspase-3 activation in silica-induced apoptosis in AMs. In addition, silica-induced lactate dehydrogenase leakage from AMs, used as an index of cell viability, was also significantly lessened in the presence of Ac-DEVD-CHO (data not shown).


View larger version (33K):
[in this window]
[in a new window]
 
Fig. 8.   Inhibitory effects of a caspase-3 inhibitor N-acetyl-Asp-Glu-Val-Asp aldehyde (Ac-DEVD-CHO) on caspase-3 (A) and silica-induced apoptosis (B). Ac-DEVD-CHO (50 µM) was pretreated for 1 h before silica exposure (100 µg for 4 h). Data are means ± SD; n = 3 experiments. Significant difference (P < 0.05) from: *control group; #silica-only group (by 1-way ANOVA with Scheffé's test).

Protective effects of ebselen against silica-induced oxidative stress, caspase-3 activation, and apoptosis in AMs. The inhibitory effects of ebselen against silica-induced ROS formation as determined by the lucigenin-dependent CL and DCFH-DA fluorescence tests are presented in Fig. 9, A and B, respectively. Ebselen not only completely abolished silica-induced O2-· formation in AMs but also markedly reduced the O2-· level in the control cells. Ebselen was also able to significantly lower the H2O2 content in silica-treated AMs, although cells treated with both silica and ebselen still had a higher level of H2O2 than the control cells. The suppressing effect of ebselen on silica-induced caspase-3 activation was also investigated, and the results are presented in Fig. 9C. It was found that ebselen markedly reduced caspase-3 activity compared with that in the silica-only group. Figure 9D shows the results of the TUNEL assay in the presence of ebselen. Ebselen significantly reduced the percentage of TUNEL-positive cells in silica-exposed AMs, whereas ebselen alone did not cause any evident effect on apoptosis. Moreover, ebselen was also able to significantly inhibit lactate dehydrogenase leakage in silica-treated AMs (data not shown).


View larger version (25K):
[in this window]
[in a new window]
 
Fig. 9.   Protective effects of ebselen against silica-induced oxidative stress and apoptosis in rat AMs. A: O2-· level as measured by lucigenin-dependent CL. B: H2O2 level as determined by DCFH-DA test. C: caspase-3 activation as analyzed by cleavage of substrate Ac-DEVD-AMC. D: apoptosis as assessed by TUNEL assay. Ac-DEVD-CHO (50 µM) was pretreated for 1 h before silica exposure (100 µg for 4 h). Data are means ± SD; n = 3 or 4 experiments. Significant difference (P < 0.05) from: * control group; # silica-only group (by 1-way ANOVA with Scheffé's test).


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

Although silica is known to be highly toxic to AMs, the form of cell death in silica-treated AMs has not been well studied. In a pioneering study, Sarih et al. (30) reported that silica induces apoptosis in macrophages, and subsequent studies (19, 20, 23) confirmed such a phenomenon under both in vivo and in vitro conditions as detected by morphological alterations and/or DNA gel electrophoresis. In the present study, silica-induced apoptosis in AMs was demonstrated by 1) a time- and dose-dependent increase in TUNEL-positive cells, 2) a time- and dose-dependent increase in sub-G1 cells, and 3) reduced cell size and increased cell granularity. Comparatively, the TUNEL assay with flow cytometry as used in this study is believed to be a specific and sensitive method for measuring DNA fragmentation, a hallmark of apoptosis quantitatively (1, 12). Results from this study therefore provide convincing evidence that apoptosis is an important form of cell death in silica-exposed AMs.

It is well known that ROS and oxidative damage are common and important mediators of apoptosis based on the following observations: 1) the addition or induction of ROS leads to apoptosis, 2) the depletion of cellular antioxidants promotes apoptosis, 3) apoptosis is blocked by antioxidants, and 4) many apoptosis-regulating proteins such as Bcl-2 and p53 act through the oxidant-antioxidant pathway (2, 6, 21, 25, 28). On the other hand, there is substantial evidence showing that ROS and oxidative stress play an important role in silica-induced pulmonary cell injury. First, silica promotes the production of various species of ROS in cell-free systems as well as in silica-exposed cells or tissues (33, 38). Results from our study also confirm that silica exposure enhances ROS production in cultured rat AMs (Fig. 1). Second, silica is able to induce oxidative damage such as lipid peroxidation and oxidative DNA damage both in vivo and in vitro (10, 34). Third, many antioxidants are capable of protecting against silica-induced cytotoxicity (18, 35). Therefore, it will be of interest to explore the involvement of ROS and oxidative stress in silica-induced apoptosis. In this study, it was noted that a significant increase in ROS formation preceded other apoptotic events such as caspase-3 activation and DNA fragmentation. Ebselen, a potent antioxidant and ROS scavenger (36, 41), was able to reduce the ROS level, inhibit caspase-3 activation, and eventually protect against silica-induced apoptosis (Fig. 9). It appears that ROS and oxidative stress may act as initiating factors in silica-induced apoptosis in cultured rat AMs.

At present, the exact signaling pathway of apoptosis triggered by ROS and oxidative stress remains to be elucidated. An important step in apoptosis is the activation of a series of proteases called caspases. Among 14 caspases identified so far, caspase-3 has been found to be one of the most important effectors in executing apoptosis (29). In the present study, the critical role of caspase-3 activation in silica-induced apoptosis was demonstrated by 1) a time- and dose-dependent increase in caspase-3 activity in silica-treated AMs, 2) evident PARP cleavage, and, more importantly, 3) the protective effect of Ac-DEVD-CHO, a specific caspase-3 inhibitor, against silica-induced apoptosis. Results obtained from this study are generally consistent with those reported previously that caspase-3 or cysteine protease protein-32 was involved in silica-induced apoptosis in human macrophages in vitro (20). Furthermore, the present study also evaluated the activation of caspase-9, an upstream caspase that is activated by the formation of apoptosome with cytochrome c, apoptotic protease-activating factor-1, and dATP (14, 24). Although no direct evidence is currently available, it is believed that the elevated level of ROS generated from silica exposure may initiate an apoptotic pathway through mitochondria, causing cytochrome c release and caspase-9 activation, which subsequently lead to caspase-3 activation, PARP cleavage, and DNA fragmentation.

On the other hand, based on the fact that both the caspase-3 inhibitor Ac-DEVD-CHO and the antioxidant ebselen failed to completely block the occurrence of apoptosis, it is believed that there are some other intermediate factors regulating the signaling pathway from ROS formation to caspase activation and DNA fragmentation. At present, it has been extensively studied that silica is able to cause nuclear factor (NF)-kappa B activation, most probably through the action of ROS (8). NF-kappa B is a very important nuclear transcription factor that is closely associated with the signaling pathway of apoptosis (4, 7). Moreover, ROS are also involved in the release of cytokines including tumor necrosis factor-alpha , interleukin-1alpha , and interleukin-1beta from silica-treated AMs (13, 30), which, in turn, are important factors in initiating or regulating apoptosis. The exact role of NF-kappa B or other NFs in silica-induced apoptosis remains to be further investigated.

At present, the biological relevance of silica-induced apoptosis to the development of pulmonary diseases such as silicosis and carcinogenesis has not been well defined. Based on the observation that crystalline silica-induced apoptosis in AMs, whereas no similar effects were found with amorphous silica or titanium dioxide, it has been proposed that the fibrotic potential of a particulate depends on its ability to cause apoptosis (19). Such a proposal was obviously supported by studies on other fibrotic agents such as bleomycin and asbestos that are able to induce apoptosis in AMs as well (15, 16). It is well known that an increase in recruitment and activation of AMs plays an important role in the pathogenesis of lung disease resulting from silica exposure (11, 26). Apoptosis has been identified to be a critical pathway for the elimination of damaged or injured cells and for the maintenance of tissue homeostasis. By studying silica-induced apoptosis in AMs and granulomatous cells under an in vivo condition, Leigh et al. (23) believed that silica-induced apoptosis has a regulatory role in the process of inflammation in silica-exposed lung tissue by the following two mechanisms: 1) attracting more AMs into the alveolar space to engulf apoptotic cells and 2) maintaining a relatively stable level of neutrophils in the inflammatory sites. On the other hand, silica-induced apoptosis may also be implied in the carcinogenic process, and the exact mechanisms involved remain to be further elucidated.

Ebselen is a synthetic heterocyclic selenoorganic compound with potent antioxidant activity. Basically, it mimics the reactions of glutathione peroxidase, with thiols as electronic donors (36). It is well known that ebselen is able to protect against oxidative damage such as lipid peroxidation both in vivo and in vitro as well as to scavenge free radicals including superoxide radicals, hydroxyl radicals, peroxyl radicals, and peroxynitrite (3, 36, 41). In the present study, ebselen was able to protect against silica-induced apoptosis in AMs, most probably through its potent antioxidant activity, by scavenging ROS formed in silica-treated AMs. Therefore, results from this study suggest that ebselen might have the potential as a preventive agent against silica-induced pulmonary injury.

In summary, results from this study suggest that apoptosis is an important form of cell death caused by silica exposure. The elevated ROS level resulting from silica exposure may act as an initiator, leading to caspase-9 and caspase-3 activation and PARP cleavage to execute the apoptotic process. Knowledge of such a signaling pathway may help to obtain a better understanding of the underlying mechanism in the development of silica-induced pathological changes.


    ACKNOWLEDGEMENTS

This study was partially supported by the China Medical Board (New York, NY) and the Centre for Environmental and Occupational Health, National University of Singapore.


    FOOTNOTES

Present address of Z. Zhang: Pathology and Physiological Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505.

Address for reprint requests and other correspondence: C. N. Ong, Dept. of Community, Occupational, and Family Medicine, Faculty of Medicine (MD3), National Univ. of Singapore, Singapore 117597, Republic of Singapore (E-mail: cofongcn{at}nus.edu.sg).

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.

Received 30 January 2000; accepted in final form 17 August 2000.


    REFERENCES
TOP
ABSTRACT
INTRODUCTION
EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
REFERENCES

1.   Allen, RT, Hunter WJ, III, and Agrawal DK. Morphological and biochemical characterization and analysis of apoptosis. J Pharmacol Toxicol Methods 37: 215-228, 1997[ISI][Medline].

2.   Anderson, KM, Seed T, Ou D, and Harris JE. Free radicals and reactive oxygen species in programmed cell death. Med Hypotheses 52: 451-463, 1999[ISI][Medline].

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

4.   Baichwal, VR, and Baeuerle PA. Apoptosis: activate NF-kappaB or die? Curr Biol 7: R94-R96, 1997[ISI][Medline].

5.   Budihardjo, I, Oliver H, Lutter M, Luo X, and Wang X. Biochemical pathways of caspase activation during apoptosis. Annu Rev Cell Dev Biol 15: 269-290, 1999[ISI][Medline].

6.   Buttke, TM, and Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today 15: 7-10, 1994[ISI][Medline].

7.   Chen, F, Castranova V, Shi X, and Demers LM. New insights into the role of nuclear factor-kappaB, a ubiquitous transcription factor in the initiation of diseases. Clin Chem 45: 7-17, 1999[Abstract/Free Full Text].

8.   Chen, F, Lu Y, Demers LM, Rojanasakul Y, Shi X, Vallyathan V, and Castranova V. Role of hydroxyl radical in silica-induced NF-kappa B activation in macrophages. Ann Clin Lab Sci 28: 1-13, 1998[Abstract].

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

10.   Daniel, LN, Mao Y, Wang TC, Markey CJ, Markey SP, Shi X, and Saffiotti U. DNA strand breakage, thymine glycol production, and hydroxyl radical generation induced by different samples of crystalline silica in vitro. Environ Res 71: 60-73, 1995[ISI][Medline].

11.   Davis, GS. Pathogenesis of silicosis: current concepts and hypothesis. Lung 164: 139-154, 1986[ISI][Medline].

12.   Gavrieli, Y, Sherman Y, and Ben-Sasson SA. Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119: 493-501, 1992[Abstract].

13.   Gossart, S, Cambon C, Orfila C, Seguelas MH, Lepert JC, Rami J, Carre P, and Pipy B. Reactive oxygen intermediates as regulators of TNF-alpha production in rat lung inflammation induced by silica. J Immunol 156: 1540-1548, 1996[Abstract].

14.   Green, DR, and Reed JC. Mitochondria and apoptosis. Science 281: 1309-1312, 1998[Abstract/Free Full Text].

15.   Hamilton, RF, Jr, Iyer LL, and Holian A. Asbestos induces apoptosis in human alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 271: L813-L819, 1996[Abstract/Free Full Text].

16.   Hamilton, RF, Jr, Li L, Felder TB, and Holian A. Bleomycin induces apoptosis in human alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 269: L318-L325, 1995[Abstract/Free Full Text].

17.   Holian, A, Kelley K, and Hamilton RF, Jr. Mechanisms associated with human alveolar macrophage stimulation by particulates. Environ Health Perspect 102, Suppl10: 69-74, 1994[ISI][Medline].

18.   Huang, SH, Leonard S, Shi X, Goins MR, and Vallyathan V. Antioxidant activity of lazaroid (U-75412E) ant its protective effects against crystalline silica-induced cytotoxicity. Free Radic Biol Med 24: 529-536, 1998[ISI][Medline].

19.   Iyer, R, Hamilton RF, Li L, and Holian A. Silica-induced apoptosis mediated via scavenger receptor in human alveolar macrophages. Toxicol Appl Pharmacol 141: 84-92, 1996[ISI][Medline].

20.   Iyer, R, and Holian A. Involvement of the ICE family of proteases in silica-induced apoptosis in human alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 273: L760-L767, 1997[Abstract/Free Full Text].

21.   Kitamura, M, and Ishikawa Y. Oxidant-induced apoptosis of glomerular cells: intracellular signaling and its intervention by bioflavinoid. Kidney Int 56: 1223-1229, 1999[ISI][Medline].

22.   Kuida, K. Caspase-9. Int J Biochem Cell Biol 32: 121-124, 2000[ISI][Medline].

23.   Leigh, J, Wang H, Bonin A, Peters M, and Ruan X. Silica-induced apoptosis in alveolar and granulomatous cells in vivo. Environ Health Perspect 105, Suppl 5: 1241-1245, 1997[ISI][Medline].

24.   Li, P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, and Wang X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91: 479-489, 1997[ISI][Medline].

25.   Mates, JM, and Sanchez-Jimenez FM. Role of reactive oxygen species in apoptosis: implications for cancer therapy. Int J Biochem Cell Biol 32: 157-170, 2000[ISI][Medline].

26.   Mossman, BT, and Churg A. Mechanisms in the pathogenesis of asbestosis and silicosis. Am J Respir Crit Care Med 157: 1666-1680, 1998[Free Full Text].

27.   Nicoletti, I, Migliorati G, Pagliacci MC, Grignani F, and Riccard C. A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry. J Immunol Methods 139: 271-279, 1991[ISI][Medline].

28.   Payne, CM, Bernsterin C, and Bernsterin H. Apoptosis overview emphasizing the role of oxidative stress, DNA damage and signal-transduction pathways. Leuk Lymphoma 19: 43-93, 1995[ISI][Medline].

29.   Porter, AG, and Janicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ 6: 99-104, 1999[ISI][Medline].

30.   Sarih, M, Souvannavong V, Brown SC, and Adam A. Silica induces apoptosis in macrophages and the release of interluekin-1alpha and interlukin-1beta . J Leukoc Biol 54: 407-413, 1993[Abstract].

31.   Shen, HM, Shi CY, Shen Y, and Ong CN. Detection of elevated reactive oxygen species level in cultured rat hepatocytes treated with aflatoxin B1. Free Radic Biol Med 21: 139-146, 1996[ISI][Medline].

32.   Shen, HM, Yang CF, and Ong CN. Induction of oxidative stress and apoptosis in sodium selenite-treated human hepatoma cells (HepG2). Int J Cancer 81: 820-828, 1999[ISI][Medline].

33.   Shi, X, Castranova V, Halliwell B, and Vallyathan V. Reactive oxygen species and silica-induced carcinogenesis. J Toxicol Environ Health B Crit Rev 1: 181-197, 1998[ISI][Medline].

34.   Shi, X, Mao Y, Daniel LN, Saffiotti U, Dalal NS, and Vallyathan V. Silica radical-induced DNA damage and lipid peroxidation. Environ Health Perspect 102, Suppl10: 149-154, 1994[ISI][Medline].

35.   Shi, X, Mao Y, Saffiotti U, Wang L, Rojanasakul Y, Leonard SS, and Vallyathan V. Antioxidant activity of tetrandrine and its inhibition of quartz-induced lipid peroxidation. J Toxicol Environ Health 46: 233-248, 1995[ISI][Medline].

36.   Sies, H. Ebselen, a selenoorganic compound as glutathione peroxidase mimic. Free Radic Biol Med 14: 313-323, 1993[ISI][Medline].

37.   Thornberry, NA. Caspases: key mediators of apoptosis. Chem Biol 5: R97-R103, 1998[ISI][Medline].

38.   Vallyathan, V, Leonard S, Kuppusamy P, Pack D, Chzhan M, Sanders SP, and Zweir J. Oxidative stress in silicosis: evidence for the enhanced clearance of free radicals from whole lungs. Mol Cell Biochem 168: 125-132, 1997[ISI][Medline].

39.   Vallyathan, V, and Shi X. The role of oxygen free radicals in occupational and environmental lung disease. Environ Health Perspect 105, Suppl1: 165-177, 1997[Medline].

40.   Wyllie, AH, Kerr JFR, and Currie AR. Cell death: the significance of apoptosis. Int Rev Cytol 68: 251-306, 1990.

41.   Yang, CF, Shen HM, and Ong CN. Protective effect of ebselen against hydrogen peroxide-induced cytotoxicity and DNA damage in HepG2 cells. Biochem Pharmacol 57: 273-279, 1999[ISI][Medline].

42.   Zhang, Z, Shen HM, Zhang QF, and Ong CN. Critical role of GSH in silica-induced oxidative stress, cytotoxicity, and genotoxicity in alveolar macrophages. Am J Physiol Lung Cell Mol Physiol 277: L743-L748, 1999[Abstract/Free Full Text].


Am J Physiol Lung Cell Mol Physiol 280(1):L10-L17
1040-0605/01 $5.00 Copyright © 2001 the American Physiological Society