Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • Subject Collections
    • JASN Podcasts
    • Archives
    • Saved Searches
    • ASN Meeting Abstracts
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Subscriptions
  • More
    • About JASN
    • Alerts
    • Advertising
    • Editorial Fellowship Program
    • Feedback
    • Reprints
    • Impact Factor
  • ASN Kidney News
  • Follow JASN on Twitter
  • Visit ASN on Facebook
  • Follow JASN on RSS
  • Community Forum
Cell and Transport Physiology
You have accessRestricted Access

Cisplatin Induces Apoptosis in LLC-PK1 Cells via Activation of Mitochondrial Pathways

Moon Soo Park, Maryely De Leon and Prasad Devarajan
JASN April 2002, 13 (4) 858-865;
Moon Soo Park
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Maryely De Leon
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Prasad Devarajan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

ABSTRACT. Cisplatin, a commonly used chemotherapeutic agent, has a major limitation because of its nephrotoxicity. Recent studies have shown that cisplatin causes apoptotic cell death in renal tubule cells, but the underlying molecular mechanisms remain to be elucidated. In this study, cisplatin was found to induce apoptosis in a dose- and duration-dependent manner in cultured proximal tubule (LLC-PK1) cells, as evidenced by DNA laddering and TdT-mediated dUTP nick end-labeling assay. Pretreatment with the specific caspase 9 inhibitor LEHD-CHO completely prevented the apoptosis, whereas the caspase 8 inhibitor IETD-fmk had no effect. Furthermore, the activity of caspase 9 was upregulated about sixfold by cisplatin in a dose-dependent manner. These results implicated the caspase 9–dependent mitochondrial apoptotic pathways. Indeed, cisplatin triggered a duration-dependent translocation of cytochrome c from the mitochondria to the cytosol, by immunofluorescence and Western blots. Cisplatin treatment also resulted in the duration-dependent activation and mitochondrial translocation of the pro-apoptotic molecule Bax, by immunofluorescence. Finally, cisplatin induced a duration-dependent onset of the mitochondrial permeability transition. Our results indicate that cisplatin induces apoptosis in LLC-PK1 cells via activation of mitochondrial signaling pathways. The sequence of events may be summarized as follows: activation of Bax induces mitochondrial permeability transition, leading to release of cytochrome c, activation of caspase 9, and entry into the execution phase of apoptosis. Inhibition of this specific pathway may provide a strategy to minimize cisplatin-induced nephrotoxicity.

Cisplatin is one of the most widely used chemotherapeutic agents for the treatment of several human malignancies. The efficacy of cisplatin is dose dependent, but the risk of nephrotoxicity frequently hinders the use of higher doses to maximize its antineoplastic effects. Nephrotoxicity after cisplatin treatment is common and may manifest after a single dose with acute renal failure or may present with a chronic syndrome of renal electrolyte wasting. Cisplatin accumulates in cells from all nephron segments but is preferentially taken up by the highly susceptible proximal tubule cells within the S3 segment, which bear the brunt of the damage (1). The cytotoxic effects of cisplatin are postulated to occur via several mechanisms, including inhibition of protein synthesis, mitochondrial injury, and DNA damage (2,3), leading ultimately to activation of programmed cell death pathways in tumor cells (4) as well as renal tubule cells (5–15).

Apoptosis or programmed cell death is characterized by distinct morphologic changes consisting of cell shrinkage, nuclear condensation, and internucleosomal DNA fragmentation. Renal tubule cell apoptosis has recently been observed in an increasing array of renal disorders (12) and is emerging as a final common pathway in response to a wide variety of cellular stresses applied at an intensity below the threshold for necrosis. This observation also holds true for cisplatin nephrotoxicity, in which necrotic cell death is encountered with higher doses whereas lower concentrations induce apoptosis (5,11). Although cisplatin-induced renal tubule cell apoptosis has been well documented, the intracellular pathways involved in the stimulus recognition, signal transduction, and execution phases of apoptosis after cisplatin exposure remain under active investigation.

In recent years, specific intracellular proteases belonging to the caspase family have emerged as crucial effectors of apoptosis (16,17). Members of this family (now totaling at least 14) are expressed as pro-enzymes and require activation by upstream stimuli to commit a cell into the execution phase of apoptosis. It is convenient to classify the major intracellular apoptotic pathways according to the type of pro-caspase that is activated (16,17). Activation of the initiator pro-caspase 8 results predominantly from signaling via integral membrane death receptors such as Fas and TNFR1 (18). On the other hand, activation of the initiator pro-caspase 9 is dependent primarily on mitochondrial signaling pathways regulated by members of the Bcl-2 family (19). Activation of pro-apoptotic Bcl-2 family members such as Bax can trigger a sequence of events that leads to alterations in mitochondrial permeability transition, release of mitochondrial cytochrome c into the cytosol, and activation of pro-caspase 9 (20–22). Once activated, both caspases 8 and 9 participate in a cascade that culminates in the activation of caspase 3, which cleaves several substrates, resulting in chromosomal DNA fragmentation and cellular morphologic changes characteristic of apoptosis (17). The anti-apoptotic Bcl-2 family members such as Bcl-2 itself play a pivotal and overriding protective role by preserving mitochondrial structure and function, preventing onset of mitochondrial permeability transition, and inhibiting the release of cytochrome c into the cytosol (20–22).

In this study, cisplatin was found to induce apoptosis in a dose- and duration-dependent manner in cultured proximal tubule (LLC-PK1) cells. Pretreatment with the specific caspase 9 inhibitor LEHD-CHO completely prevented the apoptosis, whereas the caspase 8 inhibitor IETD-fmk had no effect. Furthermore, the activity of caspase 9 was upregulated about sixfold by cisplatin in a dose-dependent manner, whereas caspase 8 activity was not significantly altered. Cisplatin also resulted in a duration-dependent translocation of cytochrome c from the mitochondria to the cytosol, activation and mitochondrial translocation of Bax, and induction of the mitochondrial permeability transition. Our results indicate that cisplatin induces apoptosis in cultured proximal tubule cells via activation of a mitochondrial signaling pathway.

Materials and Methods

Cell Culture and Cisplatin Treatment

LLC-PK1 cells (porcine proximal tubule cells, American Type Culture Collection, Rockville, MD) were cultured in plastic six-well tissue culture plates (Costar, Cambridge, MA) in minimal essential medium alpha supplemented with 10% fetal bovine serum (Life Technologies BRL, Gaithersburg, MD). Confluent cells were incubated with varying concentrations (0, 25, 50, or 100 μM) of cisplatin (Sigma, St. Louis, MO) for different time periods. For the caspase inhibitor studies, cells were pretreated with either the caspase 8 inhibitor IETD-fmk or the caspase 9 inhibitor LEHD-CHO (both from Clontech, La Jolla, CA) 1 h before cisplatin exposure.

Apoptosis Assays

Internucleosomal DNA fragmentation was detected primarily by DNA laddering assay (23). In brief, equal number of cells were resuspended in 500 μl of lysis buffer (1% sodium dodecyl sulfate, 25 mM ethylenediaminetetraacetic acid, and 1 mg/ml proteinase K [pH 8]) and incubated overnight at 50°C. Ribonuclease A (10 mg/ml) was then added for an additional 2-h incubation at 37°C. The chromosomal DNA was extracted with phenol/chloroform, precipitated with ethanol, and analyzed by agarose gel electrophoresis followed by staining with ethidium bromide to reveal the fragmentation pattern. DNA fragmentation was confirmed in situ by use of the TdT-mediated dUTP nick end-labeling (TUNEL) assay (ApoAlert DNA Fragmentation Assay Kit, Clontech), by which fluorescein-dUTP incorporation at the free ends of fragmented DNA is visualized by use of fluorescence microscopy (23). Cells grown on cover slips and subjected to control conditions or cisplatin were washed with phosphate-buffered saline (PBS) and fixed with 4% formaldehyde/PBS for 30 min at 4°C. After permeabilization with 0.2% triton X-100/PBS for 15 min at 4°C, cells were incubated with a mixture of nucleotides and TdT enzyme for 60 min at 37°C in a dark, humidified incubator. The reaction was terminated with 2× SSC, the cells washed with PBS, and the cover slips mounted on glass slides with Crystal/mount (Biomeda, Foster City, CA). Fluorescent nuclei were detected by visualization with a microscope equipped with fluorescein filters (IX70, Olympus).

Caspase Assays

Caspase 8 and caspase 9 activity assays were performed by use of the ApoAlert Caspase 8 Colorimetric Assay and the Caspase 9 Fluorescence assay kits (both from Clontech), respectively. Equal numbers of control or cisplatin-treated cells were incubated in cell lysis buffer for 10 min, centrifuged, the supernatants incubated in reaction buffer that contained IETD-AFC (specific substrate for caspase 8) or LEHD-AMC (specific substrate for caspase 9) at 37°C for 1 h. The activity was assayed by use of a spectrophotometer for caspase 8 and a fluorometer for caspase 9. The specificity of changes in caspase 9 activity was confirmed by the addition of the caspase 9 inhibitor LEHD-CHO before incubation in caspase 9 substrate, in parallel experiments.

Western Blot Analyses

For the detection of released cytochrome c, cytosolic extracts were prepared as described elsewhere (22). In brief, cells were washed with PBS, incubated for 30 min on ice in 300 μl mitochondrial buffer (68 mM sucrose, 200 mM mannitol, 50 mM KCl, 1 mM ethyleneglycol-bis[β-aminoethyl ether]-N,N′tetraacetic acid, 1 mM dithiothreitol, and 1× Complete [Roche Molecular Biochemicals, Indianapolis, IN] protease inhibitor) homogenized by six passages through a 25-guage needle, and centrifuged at 4°C at 800 × g. The supernatant was centrifuged at 4°C at 14,000 × g for 10 min and the resultant supernatant (cytosol) subjected to Western blotting. Protein concentrations were determined by the Bradford assay (Bio-Rad, Hercules, CA), and equal amounts of protein were loaded (30 μg in each lane). Monoclonal antibody to cytochrome c (clone C-7, Upstate Biotechnology, Lake Placid, NY) was used at 1:1000 dilution. Monoclonal antibody to α-tubulin (Sigma) was used at 1:10,000 dilution for confirmation of equal protein loading. Immunodetection of transferred proteins was performed by use of enhanced chemiluminescence (Amersham, Arlington Heights, IL).

Immunocytochemical Analyses

The subcellular distribution of cytochrome c and Bax in LLC-PK1 cells was determined by double labeling as described elsewhere (24). Briefly, cells grown on cover slips to confluence were incubated with 50 μM cisplatin for varying time periods, washed twice with cold 1× PBS, fixed in 4% formaldehyde on ice for 25 min, permeabilized in 0.2% Triton X-100 in PBS on ice, and blocked with goat serum at room temperature for 60 min. After a 60-min incubation simultaneously with both primary antibodies at room temperature, the cells were washed extensively with PBS and exposed simultaneously to rhodamine-conjugated goat anti-mouse (GAM-Cy3, Amersham) and fluorescein-conjugated goat anti-rabbit (GAR-Cy2, Amersham) secondary antibodies at 1:1000 dilution for 30 min in the dark. After being washed, the cover slips were mounted on glass slides with crystal mount and visualized with a microscope equipped with rhodamine and fluorescein filters. Monoclonal antibody to cytochrome c (clone C-7, Upstate Biotechnology) was used at 1:400 dilution in 2% bovine serum albumin/10% goat serum. The polyclonal antibody to the N-terminal of Bax (Bax-NT, Upstate Biotechnology) that recognizes only the activated form of Bax was used at 1:250 dilution. In a separate set of experiments, cells were double stained with polyclonal anti–Bax-NT as above, and with a monoclonal antibody against the mitochondrial voltage-dependent anion channel (Calbiochem, La Jolla, CA) at a 1:250 dilution, to confirm the mitochondrial localization of activated Bax.

Mitochondrial Permeability Transition Detection

Mitochondrial permeability transition was assessed by use of the ApoAlert Mitochondrial Membrane Sensor kit (Clontech), which contains a cationic dye (MitoSensor) that freely enters the mitochondria of normal cells and aggregates into polymers that are detected by red fluorescence. Alterations in mitochondrial membrane potential result in a cytosolic accumulation of monomeric dye, which emits a green fluorescence. Cells grown on cover slips to confluence were incubated with 50 μM cisplatin for varying time periods, rinsed with serum-free medium, and incubated for 15 min at 37°C in 1 ml of incubation buffer that contained 1 μl MitoSensor dye. After a gentle rinse with incubation buffer, the cells were examined under a fluorescence microscope.

Results

Cisplatin Induces a Dose- and Duration-Dependent Apoptosis in LLC-PK1 Cells

Cultured LLC-PK1 cells were incubated with varying concentrations of cisplatin for 24 h and subjected to two distinct apoptosis assays. In multiple experiments, internucleosomal DNA fragmentation was clearly evidenced by the characteristic 180-bp laddering pattern in cisplatin-treated cells. The DNA laddering was just detectable in 25 μM cisplatin but was clearly evident in the 50 and 100 μM cisplatin-treated cells (Figure 1A). These results indicate that the pro-apoptotic effects of cisplatin are dose-dependent, with the maximum DNA laddering seen in the 50 μM cisplatin-treated cells. We then examined the effect of varying the duration of cisplatin treatment, using the 50 μM dose. The DNA laddering was evident only after 12 h of incubation (Figure 1B), which indicates that the pro-apoptotic effects of cisplatin are also duration-dependent. The presence of apoptosis was confirmed by TUNEL assay, which revealed intense TUNEL-positive nuclei with nuclear fragmentation in the cells treated with 50 μM cisplatin for 12 or 24 h (Figure 2).

Figure1
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 1. Cisplatin (Cis) induces a dose- and duration-dependent, caspase 9–inhibitable apoptosis in LLC-PK1 cells. Cells were incubated in various concentrations of cisplatin for different periods as shown and subjected to DNA laddering assay. (A) Apoptosis was just detectable in 25 μM cisplatin but was clearly evident in the 50 and 100 μM cisplatin-treated cells. The first lane contains a 100 bp DNA marker. (B) Apoptosis was evident only after 12 h of cisplatin (50 μM) exposure. (C) Pretreatment with caspase 9 inhibitor LEHD-CHO completely abrogated cisplatin-induced apoptosis, whereas caspase 8 inhibitor IETD-fmk was devoid of a protective effect. The last lane contains a 100-bp DNA marker. This result is representative of four separate experiments.

Figure2
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 2. TdT-mediated dUTP nick end-labeling (TUNEL) assay confirms cisplatin-induced apoptosis. LLC-PK1 cells treated with cisplatin (50 μM) for varying time periods displayed intensely TUNEL-positive nuclei at the 12- and 24-h intervals, indicative of apoptosis. Arrows point to fragmented nuclei characteristic of apoptosis. This result is representative of three separate experiments. Bar, 10 μm.

Cisplatin-Induced Apoptosis Is Caspase 9–Dependent

Because it was of interest to identify the intracellular apoptotic pathways induced by cisplatin, we sought to first determine the type of pro-caspase that was activated. Pretreatment with the specific caspase 8 inhibitor IETD-fmk had no effect on cisplatin-induced DNA laddering, whereas pretreatment with the specific caspase 9 inhibitor LEHD-CHO dramatically abrogated the apoptosis (Figure 1C). Furthermore, the activity of caspase 8 was essentially unchanged in cisplatin-treated cells (Figure 3), whereas caspase 9 activity was significantly upregulated by cisplatin in a dose-dependent manner (about sixfold with the 50 μM dose), as shown in Figure 4. This increase in activity was completely prevented by addition of caspase 9 inhibitor to the assay mixture, which attests to the specificity of the activation.

Figure3
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 3. Cisplatin-induced apoptosis is not associated with a significant increase in caspase 8 activity. Cells were treated for 24 h with various concentrations of cisplatin as shown and assayed for caspase 8 activity. Values are means ± SD from three separate measurements, expressed as percentage change of control (Con).

Figure4
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 4. Cisplatin induces a dose-dependent increase in caspase 9 activity. Cells were treated for 24 h with various concentrations of cisplatin as shown and assayed for caspase 9 activity in the absence or presence (+Inh) of specific caspase 9 inhibitor. Values are means ± SD from three separate measurements, expressed as percentage change of control. *P < 0.05 versus control.

Cisplatin Induces a Duration-Dependent Translocation of Cytochrome c

The above results implicated the caspase 9–dependent mitochondrial apoptotic pathways, which we investigated first by determining the distribution of cytochrome c. Subcellular fractionation followed by Western blots showed that cytochrome c was released from the mitochondria into the cytosol in a duration-dependent fashion (Figure 5). The release of cytochrome c was evident at the 9-h incubation point with 50 μM cisplatin and appeared to be complete, with no evidence for enhanced release at further time points. The duration-dependent escape of cytochrome c into the cytosol was confirmed by immunofluorescence. Cytochrome c was detected in a punctate mitochondrial distribution in control cells and cells treated with 50 μM cisplatin for 6 h (Figure 6). However, cytochrome c assumed a diffuse cytosolic localization at the 9-h incubation point and remained cytosolic at 12 h (Figure 6).

Figure5
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 5. Cisplatin induces a duration-dependent translocation of cytochrome c (Cyto c). Cytosolic extracts from cells treated with 50 μM cisplatin for various periods as shown were analyzed by Western blot with antibodies to cytochrome c or tubulin (as a marker of equal protein loading). The release of cytochrome c was evident at the 9-h incubation point and appeared to be complete, with no evidence for enhanced release at further time points. These results were reproducible in three separate experiments.

Figure6
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 6. Cisplatin induces a duration-dependent translocation of Bax. Control cells or cells treated with 50 μM cisplatin for various periods were double-labeled with antibodies to cytochrome c and Bax-NT and visualized by immunofluorescence microscopy with rhodamine filter (to detect cytochrome c) or fluorescein filter (for activated Bax). Control cells and cells at the 6-h incubation period showed a punctate mitochondrial distribution for cytochrome c and absence of Bax activation. Treated cells at the 9- and 12-h incubation points revealed a diffuse cytosolic pattern of cytochrome c staining and a distinct punctate mitochondrial staining with Bax-NT antibody, which indicates activation and mitochondrial translocation of Bax. Cells after 9 h of cisplatin also showed a colocalization of Bax and the voltage-dependent anion channel, which confirms the mitochondrial distribution of activated Bax. Representative of three experiments. Bar, 5 μm.

Cisplatin Induces a Duration-Dependent Activation and Translocation of Bax

Because cytochrome c release can occur after activation and mitochondrial insertion of Bax, it was next of significant interest to assess the status of Bax after cisplatin exposure. We used an antibody directed to the N-terminal of Bax. The N-terminal is normally inaccessible in the inactive conformation and is exposed for antibody recognition only when activated. Accordingly, activated Bax could not be detected in control cells and cells treated with 50 μM cisplatin for 6 h (Figure 6), which indicates absence of activation. However, activated Bax was clearly evident in a punctate mitochondrial distribution at the 9- and 12-h incubation points (Figure 6). Indeed, in multiple experiments, double labeling of cells at the 9-h treatment period with antibodies to cytochrome c and Bax-NT consistently showed that every cell that was positive for cytosolic cytochrome c was also decorated by activated mitochondrial Bax (Figure 6). The mitochondrial distribution of activated Bax at the 9-h incubation point was confirmed by colocalization with voltage-dependent anion channel (not shown).

Cisplatin Induces a Duration-Dependent Mitochondrial Permeability Transition

Because one of the postulated mechanisms by which activation and mitochondrial insertion of Bax causes cytochrome c release involves changes in mitochondrial membrane potential, we assayed for mitochondrial permeability transition after cisplatin exposure using the MitoSensor probe. In control cells and cells treated with 50 μM cisplatin for 6 h, the cationic dye entered the mitochondria, aggregated into polymers, and displayed a punctate mitochondrial pattern of intense red fluorescence by use of rhodamine filters and no green fluorescence (Figure 7). In marked contrast, the majority of cells at the 9-h incubation point (and all the cells at the 12-h incubation period) were devoid of punctate red fluorescence and instead displayed a diffuse cytosolic pattern of green fluorescence when fluorescein filters were used (Figure 7). This is indicative of an inability of the cationic dye to enter the mitochondria secondary to a duration-dependent alteration in mitochondrial permeability transition, with the resultant persistence of monomeric dye in the cytoplasm.

Figure7
  • Download figure
  • Open in new tab
  • Download powerpoint

Figure 7. Cisplatin induces a duration-dependent onset of mitochondrial permeability transition. Control cells or cells treated with 50 μM cisplatin for various periods were stained with MitoSensor cationic dye and visualized by immunofluorescence microscopy. Control cells and cells at the 6-h incubation period showed a punctate mitochondrial pattern of intense red fluorescence and no green fluorescence, which indicates entry of dye into the mitochondria. In contrast, cells treated for 9 or 12 h displayed minimal red fluorescence but a marked green fluorescence in a diffuse cytosolic pattern, which indicates onset of mitochondrial permeability transition. The results were reproducible in three experiments. Bar, 10 μm.

Discussion

Cisplatin is known to cause apoptotic cell death in renal tubule cells, but the underlying mechanisms remain to be elucidated. In this study, cisplatin was found to induce apoptosis in a dose- and duration-dependent manner in cultured proximal tubule (LLC-PK1) cells. Pretreatment with the specific caspase 9 inhibitor LEHD-CHO completely prevented the apoptosis, whereas the caspase 8 inhibitor IETD-fmk had no effect. Furthermore, the activity of caspase 9 was upregulated about sixfold by cisplatin in a dose-dependent manner, whereas caspase 8 activity was not significantly altered. Cisplatin exposure resulted in a duration-dependent release of cytochrome c into the cytosol, activation and mitochondrial translocation of Bax, and onset of the mitochondrial permeability transition. Our results indicate that cisplatin induces apoptosis in LLC-PK1 cells via activation of mitochondrial pathways.

Cisplatin is one of the most widely used chemotherapeutic agents for the treatment of human malignancies. The efficacy of cisplatin is dose dependent, but nephrotoxicity frequently precludes the use of higher doses to maximize its antineoplastic effects. The renal cytotoxic effects of cisplatin may occur via the formation of DNA adducts, which can inhibit DNA replication and transcription, leading to inhibition of protein synthesis, mitochondrial injury, DNA damage, and, ultimately, to activation of programmed cell death pathways (2–4). Renal tubule cell apoptosis after cisplatin exposure has been well documented both in vivo (9,13) and in vitro (5–15). However, the intracellular pathways involved in the stimulus recognition, signal transduction, and effector phases of apoptosis after cisplatin exposure have not been fully elucidated.

In this study, cisplatin reproducibly induced apoptosis in LLC-PK1 cells in a dose- and duration-dependent manner. Apoptosis was barely detectable after a 24 h exposure to 25 μM cisplatin but was clearly evident by two distinct assays (DNA laddering and TUNEL assay) in the cells treated with the 50 μM dose. These results are in agreement with studies elsewhere that have shown that lower doses of cisplatin (8 to 100 μM range) result in apoptosis of LLC-PK1 (11,14), mouse proximal tubule (5), and human proximal tubule cells (13,25), whereas higher doses (300 to 1000 μM range) cause necrotic cell death. Also, we detected the internucleosomal DNA fragmentation only after 12 h of exposure to 50 μM cisplatin, similar to results of a recent report (11). Collectively, these results lend support to the concept that apoptosis can result from stimuli that are applied at an intensity and duration below the threshold for necrosis.

Specific proteases belonging to the caspase family are the major effectors of apoptosis, and the type of intracellular apoptotic pathway involved may be deduced from the class of pro-caspase activated, i.e., pro-caspase 9 versus 8 (16,17). Our studies demonstrate for the first time that caspase 9-dependent pathways predominate in cisplatin-induced renal cell apoptosis in vitro, because (1) caspase 9 activity was upregulated by cisplatin in a dose-dependent manner (about threefold with the 25 μM dose and sixfold with the 50 μM dose), whereas caspase 8 activity was essentially unchanged; and (2) pretreatment with the specific caspase 9 inhibitor LEHD-CHO abrogated the cisplatin-induced apoptosis, whereas caspase 8 inhibitor had no effect. It is well known that activated caspase 9 triggers a cascade that culminates in the activation of caspase 3, which cleaves several substrates, resulting in chromosomal DNA fragmentation and cellular morphologic changes characteristic of apoptosis (17). Thus our results are in agreement with studies elsewhere that have demonstrated the activation of caspase 3, the final common mediator of apoptosis in general, after cisplatin exposure (7,11,15).

Activation of caspase 9 usually occurs downstream of cytochrome c release from mitochondria (19–22). Although mitochondrial alterations in the form of dysfunction and calcium accumulation have been previously elucidated in cisplatin nephrotoxicity (2), our results demonstrate for the first time the activation of specific mitochondrial apoptotic pathways. We documented a duration-dependent activation and mitochondrial translocation of Bax, onset of mitochondrial permeability transition, and release of cytochrome c into the cytosol. All these events were noted to occur coordinately at the 9-h incubation point with 50 μM cisplatin and appeared to be complete, with no evidence for significantly enhanced occurrence at further time points. This is in full concordance with a recent study that demonstrated that the coordinate release of cytochrome c during apoptosis induced by a variety of stimuli in HeLa cells is rapid, complete, and kinetically invariant (22). Similarly, other studies have shown that cytochrome c release occurs within minutes of Bax activation (26). Our detection of DNA laddering first at the 12-h incubation point is also consistent with the induction of mitochondrial pathways at the 9-h point, because some additional time requirement would be anticipated for the activation of caspase cascades and endonucleases with resultant internucleosomal DNA cleavage. Of interest, a similar mitochondrial signaling pathway involving translocation of Bax and release of cytochrome c also results in apoptosis of cultured proximal tubule cells after an unrelated stimulus, namely hypoxia/reoxygenation injury (20). Furthermore, our result implicating activation of pro-apoptotic Bax fully complements studies elsewhere that have shown that induction or overexpression of the pivotal anti-apoptotic molecule Bcl-2 attenuates cisplatin-induced renal cell apoptosis both in vitro (6) and in vivo (10). Collectively, these results suggest that the integration of diverse pro- and anti-apoptotic signals may occur at the mitochondria and that the release of cytochrome c may decide cell fate (27).

It has been reported that cisplatin-induced apoptosis in cultured human proximal tubule cells is temporally correlated with an upregulation of the Fas/Fas ligand system (25). However, no direct cause-and-effect association was defined. Also, induction of Fas-dependent pathways would be expected to result in activation of pro-caspase 8 (18), and we found no evidence for its significant activation in our study. Thus it is likely that Fas-dependent pathways may not play a major role in cisplatin-induced renal tubule cell apoptosis in LLC-PK1 cells. However, the scenario may be quite different in a complex in vivo system such as the kidney, in which previous studies have shown activation of Fas-dependent apoptosis after ischemia (28), allograft rejection (29), and chronic renal disease (30). Indeed, activation of the Fas pathway is a common mechanism by which cytotoxic drugs such as cisplatin induce apoptosis in cell lines derived from various tumors (31). Examples include hepatoma (32), neuroblastoma (33), and colon carcinoma (34) cells. On the other hand, cisplatin-induced apoptosis has been shown to be Fas-independent in leukemic (35,36) and lung cancer (37,38) cells, and several recent studies have implicated activation of mitochondrial apoptotic pathways after cisplatin exposure of ovarian cancer (39), squamous cell carcinoma (40), melanoma (41,42), and HeLa (43) cells. Thus, it is likely that the mechanism of cisplatin-induced apoptosis may not be similar in all cells but rather may be specific to the cell type.

The mechanisms by which cisplatin activates mitochondrial apoptotic pathways remain unknown. However, a role for oxidative stress may provide an attractive hypothesis (44). Several studies have documented the importance of reactive oxygen metabolites in cisplatin-induced renal cell apoptosis (12). A variety of antioxidants attenuate cisplatin-induced apoptosis in cultured mouse (5) or human (13) proximal tubule cells, in a rat model of nephrotoxicity (10), and in a mouse model of heme oxygenase-1 gene ablation (13), resulting in improved renal function (13). Cisplatin may directly lead to the generation of reactive oxygen species (5) or may induce (via mitochondrial damage) the release of reactive oxygen molecules normally sequestered within that organelle (12). Such molecules may then trigger several of the apoptotic mechanisms identified in this study. For example, first, reactive oxygen species have been shown to promote the onset of the mitochondrial permeability transition (45). Second, oxidant stress can activate p53 (46), which in turn is a direct activator of Bax (47). Third, reactive oxidant molecules can trigger the release of cytochrome c (48) and even activate caspases (49).

In summary, we have shown that cisplatin induces a dose- and duration-dependent apoptosis in LLC-PK1 cells, via activation of mitochondrial signaling pathways. The sequence of events is as follows: activation and mitochondrial translocation of Bax leads to onset of mitochondrial permeability transition, release of cytochrome c into the cytosol, activation of caspase 9, and entry into the execution phase of apoptosis. Selective inhibition of this pathway may provide a strategy to minimize cisplatin-induced nephrotoxicity. However, in clinical practice, any attempts at altering the apoptotic response of renal cells to cisplatin must necessarily be tempered by the potential of such maneuvers to interfere with the chemotherapy of the primary tumor.

Acknowledgments

This work was supported by grants from the NIH (RO1 DK 53289) and the American Heart Association (Grant-in-Aid) to P.D.

  • © 2002 American Society of Nephrology

References

  1. ↵
    Leibbrandt MEI, Wolfgang GHI, Metz AL, Ozobia AA, Haskins JR: Critical subcellular targets of cisplatin and related platinum analogs in rat renal proximal tubule cells. Kidney Int 48: 761–770, 1995
    OpenUrlCrossRefPubMed
  2. ↵
    Brady HR, Kone BC, Stromski ME, Zeidel ML, Giebisch G, Gullans SR: Mitochondrial injury: An early event in cisplatin toxicity to renal proximal tubules. Am J Physiol (Renal Fluid Electrolyte Physiol) 258: F1181–F1187, 1990
    OpenUrlPubMed
  3. ↵
    Huang H, Zhu L, Reid BR, Drobny GP, Hopkins PB: Solution structure of a cisplatin-induced DNA interstrand cross-link. Science 270: 1842–1845, 1995
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Jiang S, Song MJ, Shin EC, Lee MO, Kim SJ, Park JH: Apoptosis in human hepatoma cell lines by chemotherapeutic drugs via Fas-dependent and Fas-independent pathways. Hepatology 29: 101–110, 1999
    OpenUrlCrossRefPubMed
  5. ↵
    Lieberthal W, Triaca V, Levine J: Mechanisms of death by cisplatin in proximal tubular epithelial cells: Apoptosis vs. necrosis. Am J Physiol (Renal Fluid Electrolyte Physiol) 270: F700–F708, 1996
    OpenUrlPubMed
  6. ↵
    Takeda M, Kobayashi M, Shirato I, Osaki T, Endou H: Cisplatin-induced apoptosis of immortalized mouse proximal tubule cells is mediated by interleukin-1 beta converting enzyme (ICE) family of proteases but inhibited by overexpression of Bcl-2. Arch Toxicol 71: 612–621, 1997
    OpenUrlCrossRefPubMed
  7. ↵
    Fukuoka K, Takeda M, Kobayashi M, Osaki T, Shirato I, Soejima A, Nagasawa T, Endou H: Distinct interleukin-1 beta-converting enzyme family proteases mediate cisplatin and staurosporine-induced apoptosis of mouse proximal tubule cells. Life Sci 62: 1125–1138, 1998
    OpenUrlCrossRefPubMed
  8. Takeda M, Kobayashi M, Shirato I, Endou H: Involvement of macromolecule synthesis, endonuclease activation and c-fos expression in cisplatin-induced apoptosis of mouse proximal tubule cells. Toxicol Lett 94: 83–92, 1998
    OpenUrlCrossRefPubMed
  9. ↵
    Megyesi J, Safirstein RL, Price PM: Induction of p21WAF1/CIP1/SDI1 in kidney tubule cells affects the course of cisplatin-induced acute renal failure. J Clin Invest 101: 777–782, 1998
    OpenUrlCrossRefPubMed
  10. ↵
    Zhou H, Miyaji T, Kato A, Fujigaki Y, Sano K, Hishida A: Attenuation of cisplatin-induced acute renal failure is associated with less apoptotic cell death. J Lab Clin Med 134: 649–658, 1999
    OpenUrlCrossRefPubMed
  11. ↵
    Lau AH: Apoptosis induced by cisplatin nephrotoxic injury. Kidney Int 56: 1295–1298, 1999
    OpenUrlCrossRefPubMed
  12. ↵
    Ueda N, Kaushal GP, Shah SV: Apoptotic mechanisms in acute renal failure. Am J Med 108: 403–415, 2000
    OpenUrlCrossRefPubMed
  13. ↵
    Shiraishi F, Curtis LM, Truong L, Poss K, Visner GA, Madsen K, Nick HS, Agarwal A: Heme oxygenase-1 gene ablation or expression modulates cisplatin-induced renal tubular apoptosis. Am J Physiol Renal Physiol 278: F726–F736, 2000
    OpenUrlCrossRefPubMed
  14. ↵
    Okuda M, Masaki K, Fukatsu S, Hashimoto Y, Inui K: Role of apoptosis in cisplatin-induced toxicity in the renal epithelial cell line LLC-PK1. Implication of the functions of apical membranes. Biochem Pharmacol 59: 195–201, 2000
    OpenUrlCrossRefPubMed
  15. ↵
    Van de Water B, Tijdens IB, Verbrugge A, Huigsloot M, Dihal AA, Stevens JL, Jaken S, Mulder GJ: Cleavage of the actin-capping protein α-adducin at Asp-Asp-Ser-Asp633-Ala by caspase-3 is preceded by its phosphorylation on serine 726 in cisplatin-induced apoptosis of renal epithelial cells. J Biol Chem 33: 25805–25813, 2000
  16. ↵
    Adams JM, Cory S: The Bcl-2 protein family: Arbiters of cell death. Science 281: 1322–1326, 1998
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Thornberry NA, Lazebnik Y. Caspases: Enemies within. Science 281: 1312–1316, 1998
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Ashkenazi A, Dixit VM: Death receptors: Signaling and modulation. Science 281: 1305–1308, 1998
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Harris MH, Thompson CB: The role of the Bcl-2 family in the regulation of outer mitochondrial membrane permeability. Cell Death Differ 7: 1182–1191, 2000
    OpenUrlCrossRefPubMed
  20. ↵
    Saikumar P, Dong Z, Weinberg JM, Venkatachalam MA: Mechanisms of cell death in hypoxia/reoxygenation injury. Oncogene 17: 3341–3349, 1998
    OpenUrlCrossRefPubMed
  21. Korsmeyer SJ, Wei MC, Saito M, Weiler S, Oh KJ, Schlesinger PH: Pro-apoptotic cascade activates BID, which oligomerizes BAK or BAX into pores that result in the release of cytochrome c. Cell Death Differ 7: 1166–1173, 2000
    OpenUrlCrossRefPubMed
  22. ↵
    Goldstein JC, Waterhouse NJ, Juin P, Evan GI, Green DR: The coordinate release of cytochrome c during apoptosis is rapid, complete and kinetically invariant. Nat Cell Biol 2: 156–162, 2000
    OpenUrlCrossRefPubMed
  23. ↵
    Feldenberg LR, Thevananther S, Del Rio M, De Leon M, Devarajan P: Partial ATP depletion induces Fas- and caspase-mediated apoptosis in MDCK cells. Am J Physiol Renal Physiol 276: F837–F846, 1999
    OpenUrlPubMed
  24. ↵
    Erkan E, De Leon M, Devarajan P: Albumin overload induces apoptosis in LLC-PK1 cells. Am J Physiol Renal Physiol 280: F1107–F1114, 2001
    OpenUrlPubMed
  25. ↵
    Razzaque MS, Koji T, Kimatori A, Taguchi T: Cisplatin-induced apoptosis in human proximal tubular epithelial cells is associated with the activation of the Fas/Fas ligand system. Histochem Cell Biol 111: 359–365, 1999
    OpenUrlCrossRefPubMed
  26. ↵
    Eskes R, Desagher S, Antonsson B, Martinou J-C: Bid induces the oligomerization and insertion of Bax into the outer mitochondrial membrane. Mol Cell Biol 20: 929–935, 2000
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Brenner C, Kroemer G: Mitochondria—the death signal integrators. Science 289: 1150–1151, 2000
    OpenUrlFREE Full Text
  28. ↵
    Nogae S, Masanobu M, Kobayashi N, Saito T, Abe K, Saito H, Nakane PK, Nakanishi Y, Koji T: Induction of apoptosis in ischemia-reperfusion model of mouse kidney: Possible involvement of Fas. J Am Soc Nephrol 9: 620–631, 1998
    OpenUrlAbstract
  29. ↵
    Matsuno T, Sasaki H, Nakagawa K, Okada Y, Endo A, Fujiwara T, Oishi M, Saito S, Yagi T, Haisa M, Tanaka N: Expression of Fas/Fas ligand and apoptosis induction during renal allograft rejection. Transplant Proc 30: 2947–2949, 1998
    OpenUrlCrossRefPubMed
  30. ↵
    Schelling JR, Nkemere N, Kopp JB, Cleveland RP: Fas-dependent fratricidal apoptosis is a mechanism of tubular epithelial cell deletion in chronic renal failure. Lab Invest 78: 813–824, 1998
    OpenUrlPubMed
  31. ↵
    Friesen C, Fulda S, Debatin KM: Cytotoxic drugs and the CD95 pathway. Leukemia 13: 1854–1858, 1999
    OpenUrlCrossRefPubMed
  32. ↵
    Muller M, Strand S, Hug H, Heinemann EM, Walczak H, Hofmann WJ, Stremmel W, Krammer PH, Galle PR: Drug-induced apoptosis in hepatoma cells is mediated by the CD95(APO-1/Fas) receptor/ligand system and involves activation of wild-type p53. J Clin Invest 99: 403–413, 1997
    OpenUrlCrossRefPubMed
  33. ↵
    Fulda S, Sieverts H, Friesen C, Herr I, Debatin KM The CD95(APO-1/Fas) system mediates drug-induced apoptosis in neuroblastoma cells. Cancer Res 57: 3823–3829, 1997
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Micheau O, Solary E, Hammann A, Dimanche-Boitrel M-T: Fas ligand-independent, FADD-mediated activation of the Fas death pathway by anticancer drugs. J Biol Chem 274: 7987–7992, 1999
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Villunger A, Egle A, Kos M, Hartmann BL, Geley S, Kofler R, Greil R: Drug-induced apoptosis is associated with enhanced Fas (Apo-1/CD95) ligand expression but occurs independently of Fas (Apo-1/CD95) signaling in human T-acute lymphatic leukemia cells. Cancer Res 57: 3331–3334, 1997
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Eischen CM, Kottke TJ, Martins LM, Basi GS, Tung JS, Earnshaw WC, Leibson PJ, Kaufmann SH: Comparison of apoptosis in wild-type and Fas-resistant cells: Chemotherapy-induced apoptosis is not dependent on Fas/Fas ligand interactions. Blood 90: 935–943, 1997
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Kawasaki M, Kuwano K, Nakanishi Y, Hagimoto N, Takamaya K, Pei XH, Maeyama T, Yoshimi M, Hara N: Analysis of Fas and Fas ligand expression and function in lung cancer cell lines. Eur J Cancer 36: 656–663, 2000
  38. ↵
    Ferreira CG, Tolis C, Span SW, Peters GJ, van Lopik T, Kummer AJ, Pinedo HM, Giaccone G: Drug-induced apoptosis in lung cancer cells is not mediated by the Fas/FasL(CD95/APO1) signaling pathway. Clin Cancer Res 6: 203–212, 2000
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Jones NA, Turner J, McIlwrath AJ, Brown R, Dive C: Cisplatin- and paclitaxel-induced apoptosis of ovarian carcinoma cells and the relationship between bax and bak up-regulation and the functional status of p53. Mol Pharmacol 53: 819–826, 1998
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Kuwahara D, Tsutsumi K, Kobayashi T, Hasunuma T, Nishioka K: Caspase-9 regulates cisplatin-induced apoptosis in human head and neck squamous cell carcinoma cells. Cancer Lett 148: 65–71, 2000
    OpenUrlCrossRefPubMed
  41. ↵
    Raffo AJ, Kim AL, Fine RL: Formation of nuclear Bax/p53 complexes is associated with chemotherapy induced apoptosis. Oncogene 19: 6216–6228, 2000
    OpenUrlCrossRefPubMed
  42. ↵
    Mandle A, Viktorsson K, Molin M, Akusjarvi G, Eguchi H, Hayashi SI, Toi M, Hansson J, Linder S, Shoshan MC: Cisplatin induces the proapoptotic conformation of Bak in a deltaMEKK1-dependent manner. Mol Cell Biol 21: 3684–3691, 2001
    OpenUrlAbstract/FREE Full Text
  43. ↵
    Horky M, Wurzer G, Kotala V, Anton M, Vojtesek B, Vacha J, Weiserska-Gadek J: Segregation of nucleolar components coincides with caspase-3 activation in cisplatin-treated HeLa cells. J Cell Sci 114: 663–670, 2001
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Baliga R, Ueda N, Walker PD, Shah SV: Oxidant mechanisms in toxic acute renal failure. Am J Kidney Dis 29: 467–477, 1997
    OpenUrl
  45. ↵
    Lemasters JJ: Mechanisms of hepatic toxicity V. Necroapoptosis and the mitochondrial permeability transition: Shared pathways to necrosis and apoptosis. Am J Physiol (Gastrointest Liver Physiol) 276: G1–G6, 1999
    OpenUrlPubMed
  46. ↵
    Chandel NS, Vander Heiden MG, Thompson CB, Schumaker PT: Redox regulation of p53 during hypoxia. Oncogene 19: 3840–3848, 2000
    OpenUrlCrossRefPubMed
  47. ↵
    Miyashita T, Reed JC: Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80: 293–299, 1995
    OpenUrlCrossRefPubMed
  48. ↵
    Reed JC: Cytochrome c: Can’t live with it—can’t live without it. Cell 91: 559–562, 1997
    OpenUrlCrossRefPubMed
  49. ↵
    Higuchi M, Honda T, Proske RJ, Yeh ETH: Regulation of reactive oxygen species-induced apoptosis and necrosis by caspase 3-like proteases. Oncogene 17: 2753–2760, 1998
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top

In this issue

Journal of the American Society of Nephrology: 13 (4)
Journal of the American Society of Nephrology
Vol. 13, Issue 4
1 Apr 2002
  • Table of Contents
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in JASN.
Enter multiple addresses on separate lines or separate them with commas.
Cisplatin Induces Apoptosis in LLC-PK1 Cells via Activation of Mitochondrial Pathways
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Cisplatin Induces Apoptosis in LLC-PK1 Cells via Activation of Mitochondrial Pathways
Moon Soo Park, Maryely De Leon, Prasad Devarajan
JASN Apr 2002, 13 (4) 858-865;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Cisplatin Induces Apoptosis in LLC-PK1 Cells via Activation of Mitochondrial Pathways
Moon Soo Park, Maryely De Leon, Prasad Devarajan
JASN Apr 2002, 13 (4) 858-865;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Role for TGF-β in Cyclosporine-Induced Modulation of Renal Epithelial Barrier Function
  • Increased Renal Responsiveness to Vasopressin and Enhanced V2 Receptor Signaling in RGS2−/− Mice
  • Impairment of Sodium Balance in Mice Deficient in Renal Principal Cell Mineralocorticoid Receptor
Show more Cell and Transport Physiology

Cited By...

  • MicroRNA-709 Mediates Acute Tubular Injury through Effects on Mitochondrial Function
  • Differences in Urinary Renal Failure Biomarkers in Cancer Patients Initially Treated with Cisplatin
  • Dichloroacetate Prevents Cisplatin-Induced Nephrotoxicity without Compromising Cisplatin Anticancer Properties
  • Role of Annexin A5 in Cisplatin-induced Toxicity in Renal Cells: MOLECULAR MECHANISM OF APOPTOSIS
  • Epoxyeicosatrienoic Acids Prevent Cisplatin-Induced Renal Apoptosis through a p38 Mitogen-Activated Protein Kinase-Regulated Mitochondrial Pathway
  • Platinum-Induced Ototoxicity in Children: A Consensus Review on Mechanisms, Predisposition, and Protection, Including a New International Society of Pediatric Oncology Boston Ototoxicity Scale
  • Cilastatin Attenuates Cisplatin-Induced Proximal Tubular Cell Damage
  • Resistance to chemotherapy and hormone therapy in endometrial cancer
  • Nutlin-3 Protects Kidney Cells during Cisplatin Therapy by Suppressing Bax/Bak Activation
  • Intracellular Delivery of the p38 Mitogen-Activated Protein Kinase Inhibitor SB202190 [4-(4-Fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)1H-imidazole] in Renal Tubular Cells: A Novel Strategy to Treat Renal Fibrosis
  • Cisplatin preferentially binds mitochondrial DNA and voltage-dependent anion channel protein in the mitochondrial membrane of head and neck squamous cell carcinoma: possible role in apoptosis.
  • Cytoprotective Effects of Hypoxia against Cisplatin-Induced Tubular Cell Apoptosis: Involvement of Mitochondrial Inhibition and p53 Suppression
  • p53-dependent Caspase-2 Activation in Mitochondrial Release of Apoptosis-inducing Factor and Its Role in Renal Tubular Epithelial Cell Injury
  • Endoplasmic Reticulum Stress-Associated Caspase 12 Mediates Cisplatin-Induced LLC-PK1 Cell Apoptosis
  • Induction of Renal Tubular Cell Apoptosis in Focal Segmental Glomerulosclerosis: Roles of Proteinuria and Fas-Dependent Pathways
  • A Novel Free Radical Scavenger, Edarabone, Protects Against Cisplatin-Induced Acute Renal Damage in Vitro and in Vivo
  • Metabolism of Cisplatin to a Nephrotoxin in Proximal Tubule Cells
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Annual Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Author Resources
  • Editorial Fellowship Program
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • JASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About JASN
  • JASN Email Alerts
  • JASN Key Impact Information
  • JASN Podcasts
  • JASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe

© 2021 American Society of Nephrology

Print ISSN - 1046-6673 Online ISSN - 1533-3450

Powered by HighWire