Abstract
Ceramides are important intermediates in the biosynthesis and degradation of sphingolipids that regulatenumerous cellular processes, including cell cycle progression, cell growth, differentiation and death. In cardiomyocytes, ceramides induce apoptosis by decreasing mitochondrial membrane potential and promoting cytochrome-c release. Ca2+ overload is a common feature of all types of cell death. The aim of this study was to determine the effect of ceramides on cytoplasmic Ca2+ levels, mitochondrial function and cardiomyocyte death. Our data show that C2-ceramide induces apoptosis and necrosis in cultured cardiomyocytes by a mechanism involving increased Ca2+ influx, mitochondrial network fragmentation and loss of the mitochondrial Ca2+ buffer capacity. These biochemical events increase cytosolic Ca2+ levels and trigger cardiomyocyte death via the activation of calpains.
Keywords: Ceramide, Metabolism, Mitochondrial dynamics, Ca2+, Cell death, Cardiomyocyte
1. Introduction
Thudichum (1884) referred to sphingolipids (from the Greek Σφιγξ: sphinx) as a peculiar group of lipids because of their enigmatic nature [1]. Ceramides represent the structural backbone common to all sphingolipids [2]. Moreover, they regulate several key cellular processes, including cell cycle, cell growth and differentiation, as well as cell death. Intracellular de novo synthesis of ceramides occurs in response to cytokines, environmental stress and chemotherapeutic agents [2]. Ceramides have also been shown to trigger different forms of cell death, depending on cell context. In this respect, it should be noted that they stimulate necrosis in lymphoid cells by increasing ROS formation and promoting ATP depletion [3].
Sphingolipid alterations occur in obesity and metabolic diseases where biological processes such as insulin sensitivity, lipid metabolism, inflammation, and immune responses are deregulated [4]. Particularly, ceramide accumulation has been implicated in the pathogenesis of multiple diseases including diabetes, cardiomyopathies and atherosclerosis [5]. More specifically, in the cardiovascular system, ceramides regulate cardiac contractility, vasomotor responses and endothelial function [6] and they also mediate cardiomyocyte apoptosis triggered by ischemia/reperfusion [7] or by TNF-α [8]. Moreover, C2-ceramide stimulates apoptosis in cardiomyocytes by reducing mitochondrial membrane potential (ψmt) and by activation of caspases 8 and 3 [9]. In addition, the anti-apoptotic effect of cardiac preconditioning may be due to reduced ceramide production during sustained ischemia in the rabbit heart [10].
Mitochondria play an important role in apoptotic cell death produced by multiple conditions [11]. C2-ceramide alters ψmt and promotes cytochrome-c (cyt-c) release and channel formation in the mitochondrial outer membrane [12,13]. In a previous study, we showed that C2-ceramide promotes a rapid fragmentation of the mitochondrial network in a concentration- and time-dependent manner and, as a consequence, the early activation of apoptosis [14]. Accordingly, C2-ceramide also stimulates mitochondrial network fragmentation in HeLa cells, which was linked to ER Ca2+ release and cell death [15].
The contribution of Ca2+ to cell death processes has been extensively described [16,17]. Ca2+ overload is a common feature of all types of cell death [17]. Several reports have shown a rise in cytoplasmic Ca2+ at both early and late stages of the apoptotic process [18]. Ca2+ release from ER and capacitative Ca2+ influx through Ca2+ release-activated Ca2+ channels have also been implicated in apoptosis [15,19,20]. In addition, ceramides cause the release of Ca2+ from the ER, increasing Ca2+ in both the cytosol, and the mitochondrial matrix. Accordingly, buffering cytoplasmic Ca2+ prevents mitochondrial damage and protects HeLa cells from apoptosis [15].
The aim of the current study was to investigate the molecular mechanisms involved in ceramide-dependent cardiomyocyte death and more specifically the role of cytoplasmic Ca2+ and mitochondria in this process. Our data show that C2-ceramide induces apoptosis and necrosis in cultured cardiomyocytes by increasing cytoplasmic Ca2+ and promoting loss of mitochondrial function.
2. Materials and methods
2.1. Reagents
Cell culture reagents, Hanks medium, Dulbecco’s modified Eagle’s medium (DMEM), M199 medium, pancreatin, gelatin, Triton X-100, 5-bromo-2′-deoxyuridine, gadolinium chloride, propidium iodide (PI), methyl pyruvate (MP), carbonyl cyanide m-chlorophenylhydrazone (CCCP), oligomycin, FITC-conjugated anti-IgG mouse antibody and the chemical inhibitors E64D, leupeptin, Z-VAD-fmk, Z-VDAD-fmk and the lactic dehydrogenase (LDH) kit were purchased from Sigma-Aldrich Co (St. Louis, MO). The sphingolipids D-erythro-sphingosine-N-acetate (C2) and dihydro-D-erythro-sphingosine-N-acetate (DH-C2), cyclosporin A (CsA) and HRP-conjugated secondary antibodies anti-IgG rabbit and mouse were from Calbiochem (La Jolla, CA). Antibodies against cytochrome-c (cyt-c) and anti-α-spectrin were from BD Pharmingen (San Diego, CA) and EMD Millipore (Billerica, MA), respectively. Anti-caspase 3 antibody was purchased in Cell Signaling (Danvers, MA). Fluo 3-AM and tetramethylrhodamine methyl ester (TMRM) were from Invitrogen (Carlsbad, CA). Organic and inorganic compounds, salts, acids and solvents were purchased from Merck (Darmstadt, Germany). CellTiter-Glo Luminescent Cell Viability Assay was purchased from Promega (Madison WI).
2.2. Animals
Rats were bred in the Animal Breeding Facility, Faculty of Chemical and Pharmaceutical Sciences, University of Chile (Santiago, Chile). Studies were approved by the Institutional Bioethical Committee, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals published by the US (NIH Publication No. 85-23, revised 1996).
2.3. Culture of cardiomyocytes
Cardiomyocytes were prepared from hearts of 1- to 3-d-old Sprague–Dawley rats as described previously [21]. Cells were maintained in DMEM/M199 (4:1) medium containing 10% FBS and serum-starved for 24 h prior stimulation. Cells were plated at a density of 1–8 × 103/mm2 on gelatin-coated 6-, 12-, 24- and 96-wells or 35-mm Petri dishes. For Ca2+ and immunofluorescence studies, cells were plated on gelatin-coated 25- and 18-mm glass coverslips in 35 mm or 12-well Petri dishes, respectively.
2.4. Cell viability and apoptosis assays
The integrity of the plasma membrane was assessed by determining the ability of cells to exclude propidium iodide (PI). The level of PI incorporation was quantified in a FACS can flow cytometer (Becton-Dickinson, San Jose, CA). Cell size was evaluated by forward-angle light scattering (FSC). PI-negative cells of normal size were considered viable [22,23]. Sub-G1 population was determined in cardiomyocytes permeabilized with methanol and labeled with propidium iodide (PI). Then the cells were analyzed using flow cytometry [22,24].
2.5. Mitochondrial membrane potential assay
Mitochondrial membrane potential (ψmt) was measured as described previously [24]. Briefly, cells were loaded with 200 nM TMRM (used in nonquenching mode) for 30 min at 37 °C. Afterwards, cells were trypsinized and fluorescence was assessed by flow cytometry (λexcitation = 543 nm; λemission = 560 nm) using a FACScan system (Becton-Dickinson, San Jose, CA,). CCCP 50 μM for 30 min was used as positive control of mitochondrial depolarization.
2.6. Intracellular and mitochondrial Ca2+ measurements
Intracellular and mitochondrial Ca2+ signals were determined as described previously [25–27]. Images were obtained from cultured cardiomyocytes preloaded with Fluo3-AM or RhodFF using an inverted confocal microscope (Carl Zeiss LSM-5, Pascal 5 Axiovert 200 microscope). Coverslips were mounted in a 1-mL capacity chamber and placed in the microscope for fluorescence measurements after excitation with a laser line of 488-nm for Fluo3-AM or 543-nm for RhodFF. The fluorescent images were collected every 0.4–2.0 s for fast signals and analyzed frame by frame with Image J software (NIH, Bethesda, MD).
2.7. Immunofluorescence studies for cytochrome-c release
Cyt-c release was evaluated by immunofluorescence as described previously [14]. Cardiomyocytes grown on coverslips were fixed with PBS containing 4% paraformaldehyde for 10 min, permeabilized with 0.3% Triton X-100 for 10 min, and blocked for 1 h with 5% BSA in PBS. Cells were then incubated with anti-cyt-c antibody at 1:400 and revealed with anti-mouse IgG-Alexa 488. Coverslips were mounted in DakoCytomation fluorescence mounting medium (DakoCytomation, Carpinteria, CA) and visualized by confocal microscope (Carl Zeiss Axiovert 135, LSM Microsystems).
2.8. Intracellular ATP levels
Cellular ATP content was measured using the luciferin/luciferase based ATP detection kit Cell Titer-Glo Luminiscent Cell Viability Assay (Promega) following the manufacturer’s instructions. Briefly, cardiomyocytes were cultured in 96-well Petri dishes and washed 3 times with PBS before incubation with the reagent. Sample luminescence was quantified in a TopCount NXT microplate luminescence counter (Perkin-Elmer, Waltham, MA). Data was normalized as fold over control.
2.9. Western blot analysis
Equal amounts of protein from cells were separated by SDS-PAGE (10% polyacrylamide gels) and electrotransfered to nitrocellulose. Membranes were blocked with 5% milk in Tris-buffered saline, pH 7.6, containing 0.1% (v/v) Tween 20 (TBST). Membranes were incubated with primary antibodies at 4 °C and re-blotted with horseradish peroxidase-linked secondary antibody [1:5000 in 1%(w/v) milk in TBST]. The bands were detected using ECL reagent followed by exposure to Kodak film and then quantified by scanning densitometry. Proteins were normalized to β-tubulin levels.
2.10. LDH release assay
Necrosis was measured by quantifying LDH activity in culture medium. LDH activity was determined by measuring the decrease in NADH (10 mg/mL) spectrophotometrically at 340 nm for 2 min, in the presence of lactate (2.5 mg/mL). The fraction of LDH released was determined by comparing LDH activity in the culture medium relative to total LDH activity.
2.11. Cardiomyocyte transfection
Small interfering RNAs (siRNA) for calpains I and II and negative control (Mission, Sigma-Aldrich Corp., St Louis, MO) were used following the manufacturer’s instructions. Cells were grown and after 48 h, transfected with siRNAs at a concentration of 25 nM with Lipofectamine™ RNAiMAX (Invitrogen, Carlsbad, CA). After 6 h incubation in Opti-mem ®, the medium was removed and replaced with 10% FBS in DMEM/M199. Calpain I/II down-regulation was assessed 48 h post-transfection. The siRNAs used for knockdown experiments were as follows:
Negative control, catalog number SIC001;
Calpain Ia, sense (5′-GUUGUGACCUUUGAUCUAU-3′), antisense (5′-AUAGAUCAAAGGUCACAAC-3′)
Calpain Ib, sense (5′-CUGACUAUGUUUGCAUGAA-3′), antisense (5′-UUCAUGCAAACAUAGUCAG-3′)
Calpain IIa, sense (5′-GAACGUACGGGAACAUUUA-3′), antisense (5′-UAAAUGUUCCCGUACGUUC-3′)
Calpain IIb, sense (5′-GUUUCAAGCUGCCCUGUCA-3′), antisense (5′-UGACAGGGCAGCUUGAAAC-3′)
Different combinations of two siRNAs against each calpain were tested, obtaining the best results with the siRNA mixture containing the specific siRNAs Ia and IIa, which was used for all the subsequent experiments (Supplementary Fig. 1A).
2.12. RNA purification and real-time reverse transcription-PCR (RT-PCR)
Total RNA was isolated using TRIzol according to the manufacturer’s protocol (Invitrogen, Carlsbad, CA). Random hexamers were used for reverse-transcription reactions using superscript II (Invitrogen, Carlsbad, CA). RT-PCR was performed using SYBR green (Invitrogen, Carlsbad, CA). Data for each transcript were normalized to both GAPDH and 18S rRNA as internal controls using the 2-ΔΔCt method. The primers used are listed below:
Calpain 1 Rat Forward 5′-TGGAGATCAGTGTCAAGGAGTTAC-3′
Calpain 1 Rat Reverse 5′-GCACTCATGCTGCCAGACTTGTCCAGGTCA-3′
Calpain 2 Rat Forward 5′-GGGCAGACCAACATCCACCTCAGCAAAAAC-3′
Calpain 2 Rat Reverse 5′-ATCTCCGCATCCTCTCCAGCCAGC-3′
2.13. Expression of results and statistical analysis
Data are presented either as means ± SD of a number (n) of independent experiments or as examples of representative experiments performed on at least three separate occasions. Data were analyzed by analysis of variance and comparisons between groups were performed using a protected Tukey t-test. A value of p < 0.05 was chosen as the limit of statistical significance.
3. Results
3.1. C2-ceramide induces apoptosis and necrosis in cardiomyocytes
Currently, there are conflicting views concerning the role of ceramides as second messengers in several cardiac pathologies [28]. One possibility to explain the death-promoting functions of ceramides invokes damage tomitochondria and alterations in calcium homeostasis. Whether suchmechanisms may be relevant in cardiomyocytes remained to be determined. Thus, we investigated the types of cell death induced by C2-ceramide in cultured rat cardiomyocytes and the role of Ca2+ and mitochondria in these events. For these experiments, we used a serum-free culture model to enhance sensitivity and detect more readily the cell death induced by C2-ceramide than previously described [14]. As shown in Fig. 1A the treatment of cardiomyocytes with C2-ceramide 20 μM during a 6 h period induces a significant increase in the sub G1 population (from 8.0% ± 1.6 to 24.2% ± 2.0), indicative of apoptosis, which however does not account for the total cell death observed under these conditions, as evaluated by PI incorporation (from 7.8% ± 0.9 to 41.4% ± 5.1). Importantly, this effect was not observed when the cells were treated with the inactive analog, dihydro- C2-ceramide, discarding a detergent effect of these short-chain lipids. The same treatment with C2-ceramide also stimulated LDH release, as determined by measuring the activity of this enzyme in the culture medium (from 10.0% ± 0.5 to 33.3% ± 6.8), as well as a decrease in the ψmt and the intracellular levels of ATP (−26.8% ± 3.4 and − 30.3% ± 2.3, using CCCP 50 μM or oligomycin 2 μM as negative and positive controls, respectively) (Fig. 1B–D). Moreover, the treatment of cardiomyocytes with C2-ceramide also induced substantial release of cyt-c after 1–3 h of treatment. This effect decreased upon exposure to C2-ceramide for longer periods of time (Fig. 1E). Collectively, these results suggest that the treatment of cardiomyocytes with C2-ceramide induces cell death by a mechanism that involved time-dependent early apoptosis followed by a delayed necrosis.
Fig. 1.
C2-ceramide induces apoptosis and necrosis in cultured cardiomyocytes. (A) Cells were treated with C2-ceramide (C2, 20 μM) or the inactive analog dihydroceramide (DH-C2) at the same concentration. After 6 h, PI incorporation and the subG1 population were quantified by FACS analysis. Values are shown as the mean ± SD of 3 independent experiments, ***P < 0.001. (B) Cells were incubated with C2-ceramide or DH-C2 (20 μM for 6 h) and necrosis was determined by the extent of LDH release to the culture medium. Data represent means ± SD (n = 3). ***P < 0.001 vs. Control (Co). (C–D) Quantification of the mitochondrial membrane potential (ψmt) and the intracellular levels of ATP in cells treated with C2-ceramide or DH-C2 (20 μM, 6 h). Values are means ± SD of n = 5 and 6 experiments, respectively, *P < 0.05, *P < 0.005 and ***P < 0.001 vs. Co. (E) Cells were incubated with C2-ceramide (C2, 20 μM) or DH-C2 for the indicated period of time and cytochrome-c was detected by indirect immunofluorescence analysis. Cytochrome-c redistribution was quantified by counting the cells with a diffuse pattern of fluorescence. (n = 7), **P < 0.01, ***P < 0.001 vs. control and ##P < 0.01 vs. C2-ceramide (C2) 3 h.
3.2. Ca2+ influx triggered by C2-ceramide stimulates cardiomyocyte death
Ceramide-induced cell death has often been associated with increments in the levels of two intracellular second messengers, Ca2+ and ROS [29–31]. To study the role of both messengers, two different approaches were used. ROS were measured by electron spin resonance (ESR) using the spin trap DMPO, whereas Ca2+ influx was assayed by confocal microscopy using the fluorescent probe Fluo 3-AM. C2-ceramide (20 μM) did not change the intracellular levels of ROS in cultured cardiomyocytes (Fig. 2A). Moreover the presence of antioxidants, including N-acetyl cysteine (NAC), Trolox (a water-soluble derivative of vitamin E) and Tiron did not reduce the C2-ceramide-induced cell death (Fig. 2B). On the other hand, ceramide increased cytoplasmic Ca2+ levels in a manner that was dependent on the presence of external Ca2+ (Fig. 3A). To investigate whether this Ca2+ signal contributes to cell death triggered by ceramide, cardiomyocytes were pretreated either with the Ca2+ chelating agent EGTA (2 mM) or with GdCl3 (10 μM), a potent but nonselective antagonist of nonselective cation currents [32]. Both agents decreased PI incorporation into the cells after treatment with C2-ceramide and also prevented decreases in ψmt and intracellular ATP levels (Fig. 3B–D), whereby Gd3+ was muchmore effective than EGTA. Additionally, both EGTA and Gd3+ delayed cyt-c redistribution, which under these conditions was no more longer significant at 3 h of treatment (Fig. 3E).
Fig. 2.
Ceramide does not stimulate ROS generation in cardiomyocytes. (A) Cells were incubated with C2-ceramide (C2, 20 μM) for a 6 h period in the presence of DMPO (200 mM) for 30 min. Cells were lysed and extracts were analyzed by electron spin resonance (ESR). Spectrometer conditions: microwave frequency 9.83 GHz, microwave power 20 mW, modulation amplitude 0.2 G, scan rate 1.25 G/s, time constant 0.5 s, and number of scans: 20. Plots are representative of 3 independent experiments (n = 3). (B) Effect of the antioxidant agents N-acetylcysteine (NAC, 5 mM), Trolox (5 mM) and Tiron (5 mM) on PI incorporation in cardiomyocytes treated with C2 ceramide (C2, 20 μM) for 6 h. (n = 3), ***P < 0.001 vs. control (Co).
Fig. 3.
Ca2+ influx induced by C2-ceramide modulates cardiomyocyte death. (A) Changes in internal cytosolic Ca2+ were investigated in individual, fluo3-AM-loaded cells maintained in medium with or without Ca2+. Cells were stimulated with C2-ceramide (C2, 20 μM) or the control (DMSO vehicle) for the time indicated by the arrow. Results are expressed as relative total fluorescence [ratio of fluorescence difference (F − Fo), to basal value (Fo)] × 100 as a function of time and are representative of 5 independent experiments in which at least 20 cells were analyzed in each case. The effect of the chelating agent EGTA (1 mM) and Gd+3 (10 μM)were studied in cells treated with C2-ceramide (C2, 20 μM) for 6 h on (B) PI incorporation, n = 3; (C)mitochondrial potential, n = 5; and(D) intracellular levels of ATP, n = 7, *P < 0.05 vs. Control (Co) and #P < 0.05 vs. C2 6 h. (E) Effect of EGTA (1 mM) and Gd+3 (10 μM) pre-incubation on the redistribution of cytochrome-c in cells treated with C2-ceramide (C2, 20 μM), n = 7, **P < 0.01, ***P < 0.001 vs. Control (Co), #P < 0.05 and ###P < 0.001 vs. C2 3 h.
3.3. Calpains but not caspases are involved in cardiomyocyte death induced by C2-ceramide
To shed light on mechanisms underlying cell death induced by C2-ceramide, activation of two important downstream effectors and key cell death mediators was evaluated in cardiomyocytes, namely the cysteine protease caspases and Ca2+-dependent calpains [33]. The activation of these proteases was evaluated by determining the cleavage of pro-caspase 3 to active caspase-3 and of α-spectrin, an important target of activated calpains [34]. As shown in the Supplementary Fig. 1B–C, under our experimental conditions only α-spectrin cleavage was detected, suggesting that calpains are activated in our experimental model. Moreover, the inhibitors of caspases Z-VDAD-fmk (50 μM) or Z-VAD-fmk (50 μM) did not prevent cell death induced by C2-ceramide, whereas the calpain inhibitors leupeptin (100 μM) or E64D (10 μM) did (Fig. 4A–B). Interestingly, leupeptin also attenuated the decrease in the intracellular levels of ATP induced by C2-ceramide. However, neither E64D nor leupeptin prevented the decrease in ψmt in cells exposed to C2-ceramide (Supplementary Fig. 2).
Fig. 4.

Calpains but not caspases are involved in cardiomyocyte death induced by C2-ceramide. PI incorporation analysis in cells treated with C2-ceramide (C2, 20 μM) for 6 h and pre-incubated (30 min) with (A) the caspase 2 (Z-VDAD-fmk) and pan-caspase (Z-VAD-fmk) inhibitors (50 μM), n = 3 or (B) the calpain inhibitors leupeptin (Leu, 100 μM) and E64D (10 μM), n = 5. ***P < 0.001 vs. Control (Co) and #P < 0.05 vs. C2 6 h. Effect of the calpain I and II knockdown (siCalpI/II) in cells treated with C2-ceramide (C2, 20 μM 6 h) on the (C) PI incorporation, n = 3; (D) mitochondrial potential, n = 3; and (E) intracellular levels of ATP, n = 3. *P < 0.05, **P < 0.01, ***P < 0.001 vs. a negative siRNA Control (siCo) and ##P < 0.01 vs. siCo C2 6 h.
Given that some calpain inhibitors are rather non-specific [35] and that calpains I and II (also known as μ-calpain and m-calpain, respectively) are both expressed in the heart requiring high Ca+2 concentration to be activated [36], both calpain isoforms were knocked down using siRNA technology. Fig. 4C shows that siRNAS against calpains I and II prevented the cell death induced by C2-ceramide. These results were similar to those obtained by the use of calpain inhibitors. Also, this strategy did not revert the decrease in the ψm and the intracellular levels of ATP (Fig. 4D–E). Altogether, these results suggest that mitochondrial dysfunction occurs prior to the onset of the calpain-dependent cell death induced by ceramides.
3.4. C2-ceramide induces cardiomyocyte death by modulating mitochondrial function
We have previously shown that C2-ceramide induced fragmentation of the mitochondrial network in cultured rat cardiomyocytes. For this reason, we evaluated here whether mitochondrial fission was triggered under our experimental conditions and whether this process was also regulated by the Ca+2 influx. Following the treatment for 6 h with C2-ceramide (20 μM) in serum-free medium, substantial mitochondrial network fragmentation was detected (Fig. 5A–B), as evidenced by an increase in the number of mitochondria per cell and a decrease in mean mitochondrial volume, both considered as criteria to document mitochondrial fission [14,37,38]. Moreover, this mitochondrial fragmentation process was prevented by the pre-incubation of cardiomyocytes with EGTA and Gd+3 (Fig. 5A–B), thereby connecting our previous results (Fig. 3) with the onset of mitochondrial dysfunction.
Fig. 5.
C2-ceramide induces cardiomyocyte death by modulating mitochondrial function. (A) Mitochondrial fragmentation induced by C2-ceramide (C2, 20 μM) was studied in cardiomyocytes pre-incubated (30 min) with or without EGTA (1 mM) and Gd+3 (10 μM). Cells were incubated with C2-ceramide for 6 h and then loaded with mitotracker green. Multi-slice imaging reconstitution was obtained by confocal microscopy to show mitochondrial morphology. The scale bar is 20 μm. (B) The mean mitochondrial volume and the number of mitochondria per cell were determined. Values shown are mean ± SD, n = 4, ***P < 0.001 vs. Control (Co) and ###P < 0.001 vs. C2 6 h. The effect of the mitochondrial transition pore inhibitor, cyclosporine (CsA, 250 nM) was studied in cells treated with C2-ceramide (C2, 20 μM) for 6 h on (C) PI incorporation, n = 3; (D) mitochondrial potential, n = 4 and; (E) intracellular levels of ATP, n = 5. **P < 0.01, ***P < 0.001 vs. Control (Co), #P < 0.05, ##P < 0.01 and ###P < 0.001 vs. C2 6 h. (F) Representative traces of mitochondrial calcium ([Ca2+]m) obtained from cardiomyocytes loaded with the Rhod-FF calcium probe, either untreated (Control) or treated with C2-ceramide (C2, 20 μM) for 6 h prior to histamine addition, which is indicated by an arrow. The mitochondrial uncoupler CCCP (50 μM) was used as a control. Results are expressed as relative total fluorescence [ratio of fluorescence difference (F − Fo), to basal value (Fo)] × 100 over time and are representative of 3 independent experiments in which at least 10 cells were analyzed in each case.
Although the relationship between mitochondrial fission and apoptosis is still controversial [39,40], our previous data [14] established a model whereby loss of ψmt precedes mitochondrial fission, which is then followed by the release of cyt-c. These results suggested that ceramide-induced mitochondrial fission is required to trigger apoptosis. Moreover, the pre-incubation of cardiomyocytes with cyclosporine A (250 nM), an inhibitor of the mitochondrial transition pore, a key step in the development of mitochondrial dependent cell death, almost completely suppressed C2-ceramide-enhanced PI incorporation, as well as decreased ψmt and intracellular ATP levels (Fig. 5C–E).
Subsequently, we sought to establish a direct relationship between the increase in cytosolic Ca2+ influx observed in cardiomyocytes treated with C2-ceramide and decreased mitochondrial Ca2+ buffer capacity, which was previously described for fragmented mitochondria [41,42]. To that end, we loaded the cells with the mitochondrial Ca2+ dye Rhod-FF and then added histamine, a classical agent that evokes a Ins(3,4,5)P3-dependent Ca2+ release from the ER [43]. As shown in Fig. 5F, histamine-stimulated mitochondrial Ca2+ uptake was significantly lower in ceramide-pretreated cardiomyocytes than controls. These results suggest that fragmented mitochondria in C2-ceramide-treated cells display decreased ER-mitochondria coupling. Moreover, these observations are indicative of reduced Ca2+ buffering capacity that may explain the increase in the cytosolic Ca2+ and subsequent activation of calpains as effectors of cell death.
Finally, it is well known that mitochondrial Ca2+ uptake stimulates mitochondrial metabolism, in terms of mitochondrial respiration and ATP production because the activities of dehydrogenases in the Krebs cycle are stimulated by Ca2+ either directly (isocitrate and α-ketoglutarate dehydrogenases) or indirectly (pyruvate dehydrogenase) [44]. If C2-ceramide causes mitochondrial fragmentation and as a consequence affects mitochondrial Ca2+ uptake and metabolism, we hypothesized that stimulating mitochondrial metabolism should decrease ceramide-induced cell death, by precluding decreased ψmt and ATP levels. To test this idea, we sought to bypass the requirement formitochondrial Ca2+ by enhancing the availability of TCA cycle intermediates and increasing the production of reducing equivalents. To this end, cells were pre-incubated with membrane-permeable methylpyruvate (MP) that is oxidized to provide reducing equivalents (NADH), which then drive oxidative phosphorylation and ATP production [45]. Fig. 6A–C shows that MP partially prevented ceramide-induced cardiomyocyte death, as assessed by PI incorporation, the decrease in the ψmt as well as the decrease in the intracellular levels of ATP.
Fig. 6.
Improved mitochondrial function prevents cardiomyocyte death induced by C2-ceramide. Effects of methyl-pyruvate (5 mM) in cells treated with C2-ceramide (C2, 20 μM) 6 h on (A) PI Incorporation, n = 5; (B) mitochondrial potential, n = 5 and; (C) intracellular levels of ATP, n = 7. *P < 0.05, ***P < 0.001 vs. Control (Co) and #P < 0.05, ##P < 0.01 and ###P < 0.001 vs. C2 6 h.
4. Discussion
Our data summarized in the graphical abstract strongly suggest that C2-ceramide induces cardiomyocyte death through a molecular mechanism involving enhanced Ca2+ influx, increased mitochondrial fragmentation and a reduction in themitochondrial Ca2+ buffer capacity. These changes lead to an increase in cytosolic Ca2+ levels, which is depicted here as responsible for the activation of calpains. Interestingly, some of these effects were partially overcome by medium supplementation with membrane-permeable methyl-pyruvate. Our data also show that C2-ceramide induces both early apoptosis and delayed necrosis. The former is dependent on Ca+2 influx (Fig. 3E) and thus can be blocked by preventing either the rise in cytoplasmic Ca2+ or loss of mitochondrial function and both approaches protect cardiomyocytes against ceramide-induced cell death.
4.1. Ceramide-induced cardiomyocyte death
A considerable body of evidence implicates ceramides in apoptotic cardiomyocyte death [28]. However, several recent reports in other cell types suggest that cell death with necrotic features may occur due to increased endogenous ceramide levels, as well as in response to treatment with cell-permeable ceramides [46–50]. In this study, we sought to define the molecular mechanisms by which cell-permeable ceramides promote cardiomyocyte death. Our current study relies heavily on the use of a flow cytometry assay that identifies different cell populations on the basis of fluorescence intensity due to DNA staining by PI that enters the cell as a consequence of augmented membrane permeability [3,46]. In order to evaluate whether cardiomyocyte membrane permeability to PI triggered by exposure to cell-permeable C2-ceramide correlated with release of cytoplasmic content from cells, LDH release, a criterion employed to identify necrotic cell death, was used in parallel. Unexpectedly as shown in Fig. 1, 6 h of treatment with C2-ceramide induced both early apoptosis and delayed necrosis. Whereas necrosis was only detected by LDH release after 6 h treatment, early apoptotic markers, such as cyt-c re-distribution were detected earlier (3 h). Ceramide-induced cell death was apparently Ca+2-dependent (Fig. 2E) and cyt-c redistribution was delayed upon addition of either EGTA or Gd3+. Consequently, we only observed decreases in ATP after 6 h of ceramide treatment, correlating with the increased LDH release, which is in agreement with the general perception that apoptosis requires ATP while necrosis occurs in its absence. Moreover, in contrast to those models where primary necrosis occurs, secondary necrosis usually follows apoptosis if the removal of apoptotic bodies is delayed or absent, as is the case of cell culture models [51]. In these cases, necrotic events, such as LDH release, occur coincident with or after cyt-c release [52], observations that strongly correlate with the results described in the present study.
4.2. Ceramide, second messengers and executors of cell death
It has been shown that ceramides increase cytoplasmic Ca2+ concentrations in different types of cells. Indeed, cytoplasmic Ca2+ levels are increased in cardiomyocytes exposed to C2-ceramide (Fig. 3A). In addition to ATP-dependent Ca2+ influx, ceramide increases Ca2+ from a thapsigargin-sensitive Ca2+ pool and triggers a subsequent capacitative Ca2+ entry in Jurkat T cells [53]. In the aforementioned studies, increased cytoplasmic Ca2+ concentrations were linked to the liberation of Ca2+ from intracellular stores and the activation of a store-operated Ca2+ channel [53]. Similarly, in our study ceramide increased cytoplasmic Ca2+ was necessary for the subsequent cardiomyocyte death because both, EGTA and Gd3+ decreased PI incorporation and also prevented the deleterious effects of ceramide on ψmt and intracellular ATP levels (Fig. 3C–D). Activation of calpains due to increased intracellular Ca2+ concentrations has been linked to cardiomyocyte death [54]. Our data agree with these observations because two different calpain inhibitors (leupeptin and E64D) and siRNAs for these proteases prevented the ceramide-induced cardiomyocyte death. However, they did not exert the same inhibitory effect on ψmt and the intracellular levels of ATP (Fig. 4). These results suggest that calpain activation represents an event that occurs downstream of mitochondrial dysfunction during cardiomyocyte death. Interestingly, caspases seem not to be involved in cardiomyocyte death induced by C2-ceramide (Fig. 4A). These results are in agreement with studies in neuroblastoma cells where synthetic ceramides induced a caspase-independent, necrosis-like morphology [55]. Moreover, ceramides kill normal lymphocytes and lymphoid cell lines in the absence of caspase activation, triggering a nonapoptotic morphology in dying cells [47]. Thus, ceramide-induced cell death is apparently not regulated by caspase activation, although the type of death triggered by these lipids is strongly dependent on the cell type and context.
4.3. Ceramide, mitochondrial morphology and function
Different studies have shown that ceramides alter the homeostasis of several organelles, including particularly mitochondria [56]. According to this evidence, C2- and C16-ceramides form channels in planar lipid bilayers allowing the release of cyt-c [56]. On the other hand, C2-ceramide also decreases ψmt in isolated heart mitochondria [57]. Our previous study established that C2-ceramide regulates mitochondrial dynamics through the stimulation of mitochondrial fission, involved among other events in the activation of cardiomyocyte apoptosis [14]. This fission process was accompanied by early translocation of the fission protein Drp-1 to the mitochondria [14]. However, the mechanisms that trigger this process remained unknown.
In the present study, we show that C2-ceramide not only induces mitochondrial fission, but also decreases ψmt and the intracellular levels of ATP (Fig. 5A–E). Also, histamine-induced mitochondrial Ca+2 uptake in cardiomyocytes is reduced (Fig. 5F), suggesting that the fragmented mitochondria observed in ceramide-treated cells display a lower Ca2+ buffering capacity. Moreover, this may explain the increase in cytosolic Ca2+ observed following C2-ceramide treatment and the subsequent activation of calpains as effectors of cell death. These data agree with previous reports establishing that a fragmented mitochondrial network affects mitochondrial Ca2+ buffering capacity and the spatial and temporal development of cytoplasmic Ca2+ signals [41]. Increases in cytosolic Ca2+ trigger the translocation of the protein Drp1 to the mitochondria, thereby unleashing mitochondrial fragmentation [58]. In our graphical abstract we propose a model where C2-ceramide induces an increase in cytosolic Ca2+, which triggers mitochondrial network fragmentation and a decrease in the function of this organelle. In this scenario, mitochondrial fragmentation reduces themitochondrial Ca+2 buffering capacity, increases cytosolic Ca2+ levels and activates calpains.
Finally, one of the most interesting findings of this study was the ability of MP to partially prevent the decrease in mitochondrial metabolism and preclude cardiomyocyte death (Fig. 6). Constitutive InsP3R-dependent Ca+2 release to mitochondria is an essential event required for efficient mitochondrial respiration and the maintenance of normal cell bioenergetics [45,59]. This process seems to be altered in cardiomyocytes treated with exogenous ceramides, given that histamine stimulation results in reduced mitochondrial Ca+2 uptake following ceramide treatment. Future work should clarify whether metabolic rescue by MP prevents the reduction in mitochondrial Ca+2 buffering capacity and also whether this may represent a beneficial approach to treat other pathological states where ceramides are implicated.
4.4. Concluding remarks
Our data provide novel evidence that cardiomyocyte mitochondria are primary targets of cell-permeable ceramides. These sphingolipids promote cytosolic Ca+2 influx, mitochondrial fragmentation and dysfunction, as well as cell death through apoptosis and necrosis. Our results also show that mitochondrial dysfunction following C2-ceramide-induced mitochondrial ψmt and ATP decrease is a major driving force for cardiomyocyte apoptosis. Thus, metabolic rescue observed upon addition of MP may represent an appropriate strategy to avoid cardiomyocyte death in a broad range of cardiac pathologies where ceramides and calpain activation may play a major role, such as in ischemia-reperfusion [36,60] and doxorubicin treatment [61,62].
Supplementary Material
Acknowledgments
This work was supported by CONICYT (grant Anillo ACT 1111 and FONDAP 1501006 to A.F.G.Q. and S.L.) and Red 120003 (to S.L. and A.F.G.Q), the National Institutes of Health (HL097768 and HL072016 to B.A.R.) and the American Heart Association (06552024 to B.A.R.). We are thankful for the PhD fellowships from CONICYT, Chile to V.P, J.K, and N.T., as well as for the FONDECYT postdoctoral funding 3110114 to R.T. V.P. holds a Postdoctoral International fellowship from Bicentennial Program, CONICYT, CHILE. We finally thank Fidel Albornoz for his excellent technical assistance.
Abbreviations
- CCCP
carbonyl cyanide m-chlorophenylhydrazone
- C2-ceramide
D-erythro-sphingosine-N-acetate
- CsA
cyclosporin A
- cyt-c
cytochrome-c
- DH-C2
dyhidro-D-erythro-sphingosine-N-acetate
- ER
endoplasmic reticulum
- LDH
lactic dehydrogenase
- MP
methyl pyruvate
- TMRM
tetramethylrhodamine methyl ester
- PI
propidium iodide
- ψmt
mitochondrial membrane potential
- TNF-α
tumor necrosis factor alpha
Footnotes
Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.bbadis.2013.04.009.
Disclosure statement
No potential conflicts of interest are reported for any of the authors.
References
- 1.Pruett ST, Bushnev A, Hagedorn K, Adiga M, Haynes CA, Sullards MC, Liotta DC, Merrill AH. Biodiversity of sphingoid bases (“sphingosines”) and related amino alcohols. J Lipid Res. 2008;49:1621–1639. doi: 10.1194/jlr.R800012-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hannun YA, Obeid LM. The ceramide-centric universe of lipid-mediated cell regulation: stress encounters of the lipid kind. J Biol Chem. 2002;277:25847–25850. doi: 10.1074/jbc.R200008200. [DOI] [PubMed] [Google Scholar]
- 3.Villena J, Henriquez M, Torres V, Moraga F, Díaz-Elizondo J, Arredondo C, Chiong M, Olea-Azar C, Stutzin A, Lavandero S, Quest AFG. Ceramide-induced formation of ROS and ATP depletion trigger necrosis in lymphoid cells. Free Radic Biol Med. 2008;44:1146–1160. doi: 10.1016/j.freeradbiomed.2007.12.017. [DOI] [PubMed] [Google Scholar]
- 4.Hla T, Dannenberg AJ. Sphingolipid signaling in metabolic disorders. Cell Metab. 2012;16:420–434. doi: 10.1016/j.cmet.2012.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Holland WL, Summers SA. Sphingolipids, insulin resistance, andmetabolic disease: newinsights fromin vivomanipulation of sphingolipidmetabolism. Endocr Rev. 2008;29:381–402. doi: 10.1210/er.2007-0025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Li X, Becker KA, Zhang Y. Ceramide in redox signaling and cardiovascular diseases. Cell Physiol Biochem. 2010;26:41–48. doi: 10.1159/000315104. [DOI] [PubMed] [Google Scholar]
- 7.Bielawska AE, Shapiro JP, Jiang L, Melkonyan HS, Piot C, Wolfe CL, Tomei LD, Hannun YA, Umansky SR. Ceramide is involved in triggering of cardiomyocyte apoptosis induced by ischemia and reperfusion. Am J Pathol. 1997;151:1257–1263. [PMC free article] [PubMed] [Google Scholar]
- 8.Krown KA, Page MT, Nguyen C, Zechner D, Gutierrez V, Comstock KL, Glembotski CC, Quintana PJ, Sabbadini RA. Tumor necrosis factor alpha-induced apoptosis in cardiac myocytes Involvement of the sphingolipid signaling cascade in cardiac cell death. J Clin Invest. 1996;98:2854–2865. doi: 10.1172/JCI119114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kong JY, Klassen SS, Rabkin SW. Ceramide activates a mitochondrial p38 mitogen-activated protein kinase: a potential mechanism for loss of mitochondrial transmembrane potential and apoptosis. Mol Cell Biochem. 2005;278:39–51. doi: 10.1007/s11010-005-1979-6. [DOI] [PubMed] [Google Scholar]
- 10.Argaud L, Prigent AF, Chalabreysse L, Loufouat J, Lagarde M, Ovize M. Ceramide in the antiapoptotic effect of ischemic preconditioning. Am J Physiol Heart Circ Physiol. 2004;286:H246–51. doi: 10.1152/ajpheart.00638.2003. [DOI] [PubMed] [Google Scholar]
- 11.Garrido C, Galluzzi L, Brunet M, Puig PE, Didelot C, Kroemer G. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 2006;13:1423–1433. doi: 10.1038/sj.cdd.4401950. [DOI] [PubMed] [Google Scholar]
- 12.Siskind LJ, Kolesnick RN, Colombini M. Ceramide forms channels inmitochondrial outer membranes at physiologically relevant concentrations. Mitochondrion. 2006;6:118–125. doi: 10.1016/j.mito.2006.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Tomassini B, Testi R. Mitochondria as sensors of sphingolipids. Biochimie. 2002;84:123–129. doi: 10.1016/s0300-9084(02)01377-9. [DOI] [PubMed] [Google Scholar]
- 14.Parra V, Eisner V, Chiong M, Criollo A, Moraga F, Garcia A, Härtel S, Jaimovich E, Zorzano A, Hidalgo C, Lavandero S. Changes inmitochondrial dynamics during ceramide-induced cardiomyocyte early apoptosis. Cardiovasc Res. 2008;77:387–397. doi: 10.1093/cvr/cvm029. [DOI] [PubMed] [Google Scholar]
- 15.Pinton P, Ferrari D, Rapizzi E, Di Virgilio F, Pozzan T, Rizzuto R. The Ca2+concentration of the endoplasmic reticulum is a key determinant of ceramide-induced apoptosis: significance for the molecular mechanism of Bcl-2 action. EMBO J. 2001;20:2690–2701. doi: 10.1093/emboj/20.11.2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Distelhorst CW, Shore GC. Bcl-2 and calcium: controversy beneath the surface. Oncogene. 2004;23:2875–2880. doi: 10.1038/sj.onc.1207519. [DOI] [PubMed] [Google Scholar]
- 17.Hanson CJ, Bootman MD, Roderick HL. Cell signalling: IP3 receptors channel calcium into cell death. Curr Biol. 2004;14:R933–5. doi: 10.1016/j.cub.2004.10.019. [DOI] [PubMed] [Google Scholar]
- 18.Distelhorst CW, Bootman MD. Bcl-2 interaction with the inositol 1,4,5-trisphosphate receptor: role in Ca(2+) signaling and disease. Cell Calcium. 2011;50:234–241. doi: 10.1016/j.ceca.2011.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jiang S, Chow SC, Nicotera P, Orrenius S. Intracellular Ca2+ signals activate apoptosis in thymocytes: studies using the Ca(2+)-ATPase inhibitor thapsigargin. Exp Cell Res. 1994;212:84–92. doi: 10.1006/excr.1994.1121. [DOI] [PubMed] [Google Scholar]
- 20.Wertz IE, Dixit VM. Characterization of calcium release-activated apoptosis of LNCaP prostate cancer cells. J Biol Chem. 2000;275:11470–11477. doi: 10.1074/jbc.275.15.11470. [DOI] [PubMed] [Google Scholar]
- 21.Foncea R, Andersson M, Ketterman A, Blakesley V, Sapag-Hagar M, Sugden PH, LeRoith D, Lavandero S. Insulin-like growth factor-I rapidly activates multiple signal transduction pathways in cultured rat cardiac myocytes. J Biol Chem. 1997;272:19115–19124. doi: 10.1074/jbc.272.31.19115. [DOI] [PubMed] [Google Scholar]
- 22.Marambio P, Toro B, Sanhueza C, Troncoso R, Parra V, Verdejo H, García L, Quiroga C, Munafo D, Díaz-Elizondo J, Bravo R, González MJ, Diaz-Araya G, Pedrozo Z, Chiong M, Colombo MI, Lavandero S. Glucose deprivation causes oxidative stress and stimulates aggresome formation and autophagy in cultured cardiac myocytes. Biochim Biophys Acta. 2010;1802:509–518. doi: 10.1016/j.bbadis.2010.02.002. [DOI] [PubMed] [Google Scholar]
- 23.Troncoso R, Vicencio JM, Parra V, Nemchenko A, Kawashima Y, del Campo A, Toro B, Battiprolu PK, Aranguiz P, Chiong M, Yakar S, Gillette TG, Hill JA, Abel ED, LeRoith D, Lavandero S. Energy-preserving effects of IGF-1 antagonize starvation-induced cardiac autophagy. Cardiovasc Res. 2012;93:320–329. doi: 10.1093/cvr/cvr321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Munoz JP, Chiong M, García L, Troncoso R, Toro B, Pedrozo Z, Díaz-Elizondo J, Salas D, Parra V, Núñez MT, Hidalgo C, Lavandero S. Iron induces protection and necrosis in cultured cardiomyocytes: role of reactive oxygen species and nitric oxide. Free Radic Biol Med. 2010;48:526–534. doi: 10.1016/j.freeradbiomed.2009.11.017. [DOI] [PubMed] [Google Scholar]
- 25.Ibarra C, Estrada M, Carrasco L, Chiong M, Liberona JL, Cárdenas C, Díaz-Araya G, Jaimovich E, Lavandero S. Insulin-like growth factor-1 induces an inositol 1,4,5-trisphosphate-dependent increase in nuclear and cytosolic calcium in cultured rat cardiac myocytes. J Biol Chem. 2004;279:7554–7565. doi: 10.1074/jbc.M311604200. [DOI] [PubMed] [Google Scholar]
- 26.Rojas-Rivera D, Díaz-Elizondo J, Parra V, Salas D, Contreras A, Toro B, Chiong M, Olea-Azar C, Lavandero S. Regulatory volume decrease in cardiomyocytes is modulated by calcium influx and reactive oxygen species. FEBS Lett. 2009;583:3485–3492. doi: 10.1016/j.febslet.2009.10.003. [DOI] [PubMed] [Google Scholar]
- 27.Chiong M, Wang ZV, Pedrozo Z, Cao DJ, Troncoso R, Ibacache M, Criollo A, Nemchenko A, Hill JA, Lavandero S. Cardiomyocyte death: mechanisms and translational implications. Cell Death Dis. 2011;2:e244. doi: 10.1038/cddis.2011.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brindley DN, Kok BPC, Kienesberger PC, Lehner R, Dyck JRB. Shedding light on the enigma of myocardial lipotoxicity: the involvement of known and putative regulators of fatty acid storage and mobilization. Am J Physiol Endocrinol Metab. 2010;298:E897–908. doi: 10.1152/ajpendo.00509.2009. [DOI] [PubMed] [Google Scholar]
- 29.Di Paola M, Zaccagnino P, Montedoro G, Cocco T, Lorusso M. Ceramide induces release of pro-apoptotic proteins frommitochondria by either a Ca2+-dependent or a Ca2+-independent mechanism. J Bioenerg Biomembr. 2004;36:165–170. doi: 10.1023/b:jobb.0000023619.97392.0c. [DOI] [PubMed] [Google Scholar]
- 30.Kong D, Xu L, Yu Y, Zhu W, Andrews DW, Yoon Y, Kuo TH. Regulation of Ca2+-induced permeability transition by Bcl-2 is antagonized by Drpl and hFis1. Mol Cell Biochem. 2005;272:187–199. doi: 10.1007/s11010-005-7323-3. [DOI] [PubMed] [Google Scholar]
- 31.Birbes H, El Bawab S, Obeid LM, Hannun YA. Mitochondria and ceramide: intertwined roles in regulation of apoptosis. Adv Enzyme Regul. 2002;42:113–129. doi: 10.1016/s0065-2571(01)00026-7. [DOI] [PubMed] [Google Scholar]
- 32.Rose RA, Hatano N, Ohya S, Imaizumi Y, Giles WR. C-type natriuretic peptide activates a non-selective cation current in acutely isolated rat cardiac fibroblasts via natriuretic peptide C receptor-mediated signalling. J Physiol. 2007;580:255–274. doi: 10.1113/jphysiol.2006.120832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chan SL, Mattson MP. Caspase and calpain substrates: roles in synaptic plasticity and cell death. J Neurosci Res. 1999;58:167–190. [PubMed] [Google Scholar]
- 34.Pedrozo Z, Torrealba N, Fernández C, Gatica D, Toro B, Quiroga C, Rodriguez AE, Sánchez G, Gillette TG, Hill JA, Donoso P, Lavandero S. Cardiomyocyte ryanodine receptor degradation by chaperone-mediated autophagy. Cardiovasc Res. 2013 doi: 10.1093/cvr/cvt029. http://dx.doi.org/10.1093/cvr/cvt029. [DOI] [PMC free article] [PubMed]
- 35.Ali MAM, Stepanko A, Fan X, Holt A, Schulz R. Calpain inhibitors exhibit matrix metalloproteinase-2 inhibitory activity. Biochem Biophys Res Commun. 2012;423:1–5. doi: 10.1016/j.bbrc.2012.05.005. [DOI] [PubMed] [Google Scholar]
- 36.Inserte J, Hernando V, Garcia-Dorado D. Contribution of calpains to myocardial ischaemia/reperfusion injury. Cardiovasc Res. 2012;96:23–31. doi: 10.1093/cvr/cvs232. [DOI] [PubMed] [Google Scholar]
- 37.Szabadkai G, Simoni AM, Chami M, Wieckowski MR, Youle RJ, Rizzuto R. Drp-1-dependent division of the mitochondrial network blocks intraorganellar Ca2+ waves and protects against Ca2+-mediated apoptosis. Mol Cell. 2004;16:59–68. doi: 10.1016/j.molcel.2004.09.026. [DOI] [PubMed] [Google Scholar]
- 38.Yu T, Robotham JL, Yoon Y. Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc Natl Acad Sci U S A. 2006;103:2653–2658. doi: 10.1073/pnas.0511154103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Martinou JC, Youle RJ. Which came first the cytochrome c release or the mitochondrial fission? Cell Death Differ. 2006;13:1291–1295. doi: 10.1038/sj.cdd.4401985. [DOI] [PubMed] [Google Scholar]
- 40.Arnoult D. Mitochondrial fragmentation in apoptosis. Trends Cell Biol. 2007;17:6–12. doi: 10.1016/j.tcb.2006.11.001. [DOI] [PubMed] [Google Scholar]
- 41.Eisner V, Parra V, Lavandero S, Hidalgo C, Jaimovich E. Mitochondria fine-tune the slow Ca(2+) transients induced by electrical stimulation of skeletal myotubes. Cell Calcium. 2010;48:358–370. doi: 10.1016/j.ceca.2010.11.001. [DOI] [PubMed] [Google Scholar]
- 42.Fülöp L, Szanda G, Enyedi B, Varnai P, Spät A. The effect of OPA1 onmitochondrial Ca2+ signaling. PLoS One. 2011;6:e25199. doi: 10.1371/journal.pone.0025199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bravo R, Vicencio JM, Parra V, Troncoso R, Munoz JP, Bui M, Quiroga C, Rodriguez AE, Verdejo HE, Ferreira J, Iglewski M, Chiong M, Simmen T, Zorzano A, Hill JA, Rothermel BA, Szabadkai G, Lavandero S. Increased ER-mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J Cell Sci. 2011;124:2143–2152. doi: 10.1242/jcs.080762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Decuypere JP, Monaco G, Bultynck G, Missiaen L, De Smedt H, Parys JB. The IP(3) receptor-mitochondria connection in apoptosis and autophagy. Biochim Biophys Acta. 2011;1813:1003–1013. doi: 10.1016/j.bbamcr.2010.11.023. [DOI] [PubMed] [Google Scholar]
- 45.Cárdenas C, Miller RA, Smith I, Bui T, Molgó J, Müller M, Vais H, Cheung KH, Yang J, Parker I, Thompson CB, Birnbaum MJ, Hallows KR, Foskett JK. Essential regulation of cell bioenergetics by constitutive InsP3 receptor Ca2+ transfer to mitochondria. Cell. 2010;142:270–283. doi: 10.1016/j.cell.2010.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hetz CA, Hunn M, Rojas P, Torres V, Leyton L, Quest AFG. Caspase-dependent initiation of apoptosis and necrosis by the Fas receptor in lymphoid cells: onset of necrosis is associated with delayed ceramide increase. J Cell Sci. 2002;115:4671–4683. doi: 10.1242/jcs.00153. [DOI] [PubMed] [Google Scholar]
- 47.Mengubas K, Fahey AA, Lewin J, Mehta AB, Hoffbrand AV, Wickremasinghe RG. Killing of T lymphocytes by synthetic ceramide is by a nonapoptotic mechanism and is abrogated following mitogenic activation. Exp Cell Res. 1999;249:116–122. doi: 10.1006/excr.1999.4474. [DOI] [PubMed] [Google Scholar]
- 48.Thon L, Möhlig H, Mathieu S, Lange A, Bulanova E, Winoto-Morbach S, Schütze S, Bulfone-Paus S, Adam D. Ceramide mediates caspase-independent programmed cell death. FASEB J. 2005;19:1945–1956. doi: 10.1096/fj.05-3726com. [DOI] [PubMed] [Google Scholar]
- 49.Kim WH, Choi CH, Kang SK, Kwon CH, Kim YK. Ceramide induces non-apoptotic cell death in human glioma cells. Neurochem Res. 2005;30:969–979. doi: 10.1007/s11064-005-6223-y. [DOI] [PubMed] [Google Scholar]
- 50.Granot T, Milhas D, Carpentier S, Dagan A, Segui B, Gatt S, Levade T. Caspase-dependent and -independent cell death of Jurkat human leukemia cells induced by novel synthetic ceramide analogs. Leukemia. 2006;20:392–399. doi: 10.1038/sj.leu.2404084. [DOI] [PubMed] [Google Scholar]
- 51.Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and immunity. Cell. 2010;140:798–804. doi: 10.1016/j.cell.2010.02.015. [DOI] [PubMed] [Google Scholar]
- 52.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. 2000;2:156–162. doi: 10.1038/35004029. [DOI] [PubMed] [Google Scholar]
- 53.Colina C, Flores A, Rojas H, Acosta A, Castillo C, Garrido MDR, Israel A, DiPolo R, Benaim G. Ceramide increase cytoplasmic Ca2+ concentration in Jurkat T cells by liberation of calcium from intracellular stores and activation of a store-operated calcium channel. Arch Biochem Biophys. 2005;436:333–345. doi: 10.1016/j.abb.2005.02.014. [DOI] [PubMed] [Google Scholar]
- 54.Bajaj G, Sharma RK. TNF-alpha-mediated cardiomyocyte apoptosis involves caspase-12 and calpain. Biochem Biophys Res Commun. 2006;345:1558–1564. doi: 10.1016/j.bbrc.2006.05.059. [DOI] [PubMed] [Google Scholar]
- 55.Ramos B, Lahti JM, Claro E, Jackowski S. Prevalence of necrosis in C2-ceramide-induced cytotoxicity in NB16 neuroblastoma cells. Mol Pharmacol. 2003;64:502–511. doi: 10.1124/mol.64.2.502. [DOI] [PubMed] [Google Scholar]
- 56.Van Blitterswijk W, Van Der Luit A. Ceramide: second messenger or modulator of membrane structure and dynamics? Biochem J. 2003;369:199–211. doi: 10.1042/BJ20021528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Di Paola M, Cocco T, Lorusso M. Ceramide interaction with the respiratory chain of heart mitochondria. Biochemistry. 2000;39:6660–6668. doi: 10.1021/bi9924415. [DOI] [PubMed] [Google Scholar]
- 58.Santel A, Frank S. Shaping mitochondria: the complex posttranslational regulation of the mitochondrial fission protein DRP1. IUBMB Life. 2008;60:448–455. doi: 10.1002/iub.71. [DOI] [PubMed] [Google Scholar]
- 59.Cárdenas C, Foskett JK. Mitochondrial Ca(2+) signals in autophagy. Cell Calcium. 2012;52:44–51. doi: 10.1016/j.ceca.2012.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Novgorodov SA, Gudz TI. Ceramide and mitochondria in ischemia/reperfusion. J Cardiovasc Pharmacol. 2009;53:198–208. doi: 10.1097/FJC.0b013e31819b52d5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lim CC, Zuppinger C, Guo X, Kuster GM, Helmes M, Eppenberger HM, Suter TM, Liao R, Sawyer DB. Anthracyclines induce calpain-dependent titin proteolysis and necrosis in cardiomyocytes. J Biol Chem. 2004;279:8290–8299. doi: 10.1074/jbc.M308033200. [DOI] [PubMed] [Google Scholar]
- 62.Delpy E, Hatem SN, Andrieu N, de Vaumas C, Henaff M, Rücker-Martin C, Jaffrézou JP, Laurent G, Levade T, Mercadier JJ. Doxorubicin induces slow ceramide accumulation and late apoptosis in cultured adult rat ventricular myocytes. Cardiovasc Res. 1999;43:398–407. doi: 10.1016/s0008-6363(99)00142-x. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





