Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Molecular Biology, Pathobiology, and Genetics

Cyclophilin D, a Component of the Permeability Transition-Pore, Is an Apoptosis Repressor

Alexis Schubert and Stefan Grimm
Alexis Schubert
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stefan Grimm
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-03-0476 Published January 2004
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The permeability transition (PT)-pore is an important proapoptotic protein complex in mitochondria. Although it is activated by many signals for apoptosis induction, the role of its various subunits in cell death induction has remained largely unknown. We found that of its components, only the voltage-dependent anion channel in the outer mitochondrial membrane and the adenine nucleotide translocator-1 (ANT-1), a PT-pore subunit of the inner membrane, are apoptosis inducers. We also report that ANT-1’s direct interactor, cyclophilin D, can specifically repress ANT-1-induced apoptosis. In addition, cotransfection experiments revealed that for a diverse range of apoptosis inducers, cyclophilin D shows the same repression profile as the compound bongkrekic acid, a specific inhibitor of the PT-pore. This activity seems to be independent of its chaperone activity, the only known function of cyclophilin D to date. Importantly, cyclophilin D is specifically up-regulated in human tumors of the breast, ovary, and uterus, suggesting that inhibition of the PT-pore via up-regulation of cyclophilin D plays a role in tumorigenesis.

INTRODUCTION

Apoptosis, or controlled cell death, is a complex process whose regulation requires the cell to integrate a variety of different signals, both pro- and antiapoptotic. In particular, mitochondria have recently been described to be integrators of such signaling pathways (1) . The permeability transition (PT)-pore is a mitochondrial protein complex on which many signals for cell death converge (2 , 3) . Because it is regulating cell death by activating downstream effectors, this protein aggregate can be regarded as a sensor for apoptosis. The PT-pore connects the inner mitochondrial membrane with the outer mitochondrial membrane. Whereas a molecular structure of this complex is still missing, biochemical data suggest that it is composed of a multitude of different components (4) . The known constituents of the PT-pore are the voltage-dependent anion channel (VDAC) and the peripheral benzodiazepine receptor in the outer mitochondrial membrane. Two reports have also implicated the proapoptotic Bcl-2-protein Bax to be part of of this complex in the outer membrane (5 , 6) . Furthermore, creatine kinase is described to associate with PT-pore subunits in the intermembrane space. The inner mitochondrial membrane harbors the PT-pore component adenine nucleotide translocator-1 (ANT-1), which is a transport protein for ADP and ATP. Cyclophilin D, a member of the cyclophilin family of chaperones, binds directly to ANT-1. In addition to these known subunits, the PT-pore could comprise further components, possibly explaining why a diverse range of cell death stimuli can activate this protein complex (4) . However, there seem to exist some proteins, most prominently among them the proapoptotic Bax protein (7) , that appear to be able to cause cell death without recruitment of the PT-pore.

The point of no return for cell death induction by mitochondria seems to be the disintegration of the outer membrane of these organelles and the release of proapoptotic factors such as apoptosis-inducing factor (8) , Smac/Diablo (9 , 10) , or cytochrome c (11) . This can be achieved by converting the PT-pore into a nonspecific pore that leads to osmotic swelling of the inner mitochondrial membrane and to the rupture of the outer membrane, providing the gate for the release of proapoptotic proteins from mitochondria (1) . However, a molecular description of how the PT-pore is activated for apoptosis is still lacking. In particular, the respective functions of the components of this protein complex for apoptosis induction or repression have not been determined.

The repression of apoptosis is a hallmark of tumorigenesis (12) . In line with this, the PT-pore has been implicated in the regulation of apoptosis in tumors. Most of these reports describe the PT-pore as a target for anticancer drugs (13) . Moreover, some associated proteins of the PT-pore such as Bax and Bcl-2 have been implicated as tumor suppressor proteins and oncogenes, respectively (14 , 15) .

The dominant activity of genes to induce or inhibit apoptosis on ectopic expression very often correlates with the function of their endogenous proteins to mediate or repress a cell death signal. It is possible that this is due to the fact that mostly protein-protein interactions are responsible for mediating the apoptosis signal within the cell. Based on this assumption, we have established a screen for apoptosis-inducing genes that are activated on overexpression (16) . In a previous report, we have described the apoptosis-inducing activity of ANT-1 (17) that was isolated using this screen. In line with our original hypothesis, ANT-1 has been shown to mediate apoptosis induction via the PT-pore (18) . In addition, this gene is dramatically up-regulated in dilated cardiomyopathy, a prevalent degenerative heart disease (19) that is characterized by an excess of apoptosis in myocytes (20) . Further investigation of ANT-1-induced apoptosis could therefore become relevant for the treatment of this fatal disease. In the present study, we have continued the molecular analysis of ANT-1-induced cell death in the context of the PT-pore and found that cyclophilin D is a repressor of apoptosis that is up-regulated in hormone-dependent human cancers.

MATERIALS AND METHODS

Materials.

All fine chemicals were from Sigma, unless otherwise specified. The pan-caspase inhibitor N-benzylcarboxy-Val-Ala-Asp-fluoromethylketone (zVAD-fmk) was obtained from Enzyme Systems.

Cell Culture.

Human embryonic kidney cells (293T) and HeLa cells were kept in DMEM (Sigma) supplemented with 5% and 10% FCS, respectively. For the generation of HeLa cells deficient in mitochondrial DNA (HeLa ρ0 cells), cells were incubated for 2 months in 100 μg/ml ethidium bromide in medium supplemented with 50 μg/ml uridine and 100 μg/ml pyruvate (21) . 293T, HeLa, and HeLa ρ0 cells were transfected with the calcium phosphate coprecipitation method as described previously (17) .

Constructs.

The cDNAs of the peripheral benzodiazipine receptor, caspase-8, ANT-2, cyclophilin D, VDAC-I, VDAC-II, creatine kinase, and the mitochondrial chaperones p10 and p60 were isolated by PCR and subcloned in the mammalian expression vector pcDNA3 (Invitrogen). They were all control sequenced. The expression construct for RIP has been described previously (22) . The caspase-2 vector was a gift from S. Kumar. The double point mutant of cyclophilin D (CyloD*R97A/H168Q) was generated by PCR using the Pwo polymerase, which has proofreading activity and was control sequenced.

Apoptosis Detection.

Apoptosis was quantified by determining the amount of sub-G1 DNA with flow cytometry [FACSCalibur; Becton Dickinson (23)] . Briefly, cells were trypsinized, washed, and resuspended in PBS. After the addition of 1.5 volumes of hypotonic propidium iodide buffer [20 μg/ml propidium iodide, 0.1% (w/v) sodium citrate, and 0.1% Triton in PBS], fluorescence-activated cell-sorting analysis was performed. A cotransfected green fluorescent protein (GFP) expression plasmid (pLantern; Life Technologies, Inc.) was used to assess the transfection efficiency. The apoptotic population was measured in relation to the percentage of green fluorescent protein-positive cells. Each condition was tested in at least three independent experiments.

Immunoblotting.

For detecting protein expression, cells were harvested by trypsinization, washed with PBS, and lysed in Triton X buffer [50 mm Tris-HCl (pH 8.0), 150 mm NaCl, and 1% Triton X-100] for 10 min on ice. After centrifugation at 14,000 × g in a microfuge, supernatants were obtained as cytoplasmic extracts that were quantified for protein content using the Bradford reagent (Bio-Rad). Equal amounts of protein were loaded on a 15% SDS-polyacrylamide gel after boiling with 1 volume of 2× sample buffer [100 mm Tris-HCl (pH 6.8), 4% SDS, 10% mercaptoethanol, 20% glycerol, and 0.05% bromphenol blue], separated in an electric field, and transferred to a polyvinylidene difluoride membrane in a semidry blotting device. The membrane was blocked for 1 h with 5% dry milk powder in Tris-buffered saline-Tween [10 mm Tris-HCl (pH 8.0), 150 mm NaCl, and 0.05% Tween] and probed overnight at 4°C with either rat monoclonal anti-hemagglutinin high-affinity antibodies (Roche), cyclophilin D (Dianova, Hamburg, Germany), or Tim23 (BD Biosciences, Heidelberg, Germany). Membranes were washed three times with Tris-buffered saline-Tween and then probed for 1 h at room temperature with secondary horseradish peroxidase-conjugated antibodies (Roche). After washing with Tris-buffered saline-Tween, the blot was developed using Super Signal enhanced chemiluminescence reagents (Pierce) and by exposure to BioMax MR films (Kodak).

Northern Hybridization.

For hybridization of the Matched Tumor/Normal Profiling Array (Clontech), the coding sequence of cyclophilin D was excised from the expression plasmid and labeled with 5′-[α-32P]dCTP using the RediPrime random prime labeling kit (Amersham) with 200 ng of plasmid and 50 μCi of [32P]dCTP. The blots were prehybridized for 3 h at 65°C in 40 ml of hybridization buffer (6× SSC, 5× Denhardt’s reagent, 0.5% SDS, and 0.1 mg/ml salmon sperm DNA), 10 ml of a 50% dextrane sulfate solution were added, and, finally, the labeled probe (approximately 3 × 107 cpm) was added to the hybridization solution and incubated at 62°C for 6 h. The blot was washed with buffer 1 (2× SSC and 0.5% SDS), buffer 2 (1× SSC and 0.5% SDS), and buffer 3 (0.5× SSC and 0.5% SDS). The blot was exposed in a BAS 2500 phosphorimager screen (Fuji-Films, Heidelberg, Germany), and the signals were analyzed and quantified using Image Gauge V3.01 software.

ANT-1 Transport Activity Measurement.

The ADP/ATP transport activity of ANT-1 was measured 24 h after transfecting ANT-1 (or β-galactosidase as a control) by a modified protocol (24) . Briefly, the cells were permeabilized with 12.5 μm digitonin for 4 min, washed, and resuspended in a buffer containing 125 μm ADP to stimulate the antiport of ATP. The ATP that was released into the supernatant was then determined by an enzymatic reaction (25) .

Detection of Mitochondrial Membrane Potential (ΔΨ).

The mitochondrial membrane potential was assayed by 3,3′-dihexyloxacarbocyanine iodide (Molecular Probes Inc.) Briefly, the transiently transfected cells were harvested and incubated with 0.5 nm 3,3′-dihexyloxacarbocyanine iodide and 2.5 μm ionomycin and analyzed by flow cytometry (FACSCalibur; Becton Dickinson).

RESULTS

Characterization of ANT-1-Induced Apoptosis.

To assess the contribution of the various PT-pore subunits for apoptosis induction, we isolated the known components of the PT-pore by reverse transcription-PCR and transfected them into 293T cells. A quantitative fluorescence-activated cell-sorting analysis revealed that ANT-1 could potently induce apoptosis (Fig. 1A) ⇓ . VDAC-I and VDAC-II, two components of the outer mitochondrial membrane, could also cause cell death but did so less efficiently. In contrast, the other subunits of this proapoptotic protein complex remained inactive for cell death induction. A Western blot showed the efficient expression of the various PT-pore components (data not shown). The difference between ANT-1, a powerful cell death inducer, and ANT-2, a gene completely inactive for cell death induction, was especially striking, given the 90% identity of these two proteins. It is therefore unlikely that nonspecific reactions such as the misfolding of ANT-1 proteins could be responsible for apoptosis induction. Furthermore, after ANT-1 transfection, we observed a 28% increase in the transport of ATP after antiport with ADP (data not shown) over control-transfected cells, suggesting that the ANT-1 protein is correctly folded in the membrane. The different behavior of ANT-1 and ANT-2 facilitated delineation of the domain(s) necessary for cell death induction in ANT-1. To this end, we generated a number of swap mutants that replaced progressively longer sequences beginning at the NH2 terminus of ANT-1 by the corresponding domains of ANT-2. After transfection, apoptosis was quantified and compared. Fig. 1B ⇓ reveals that four swap mutants (Ex 17, Ex 107, Ex 170, and Ex 207) led to a drop in apoptosis induction when compared with their preceding mutants. According to its proposed structure (26) ANT-1 traverses the membrane six times and thereby forms five intermittent loops. Accordingly, the NH2 terminus and the COOH terminus are proposed to protrude into the intermembrane space of mitochondria. The mapping experiment therefore characterizes four domains in ANT-1 that contribute to apoptosis induction: the very NH2 terminus; and the second, third, and fourth loop. Of note, the exchange of the third, matrix-restricted loop led to the biggest decline in cell death induction.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Characterization of adenine nucleotide translocator-1 (ANT-1)-induced apoptosis. A, test of the various components of the permeability transition-pore for apoptosis induction. Five μg of the respective expression plasmids (adenine nucleotide translocator-2, ANT-2; voltage-dependent anion channel-1, VDAC I; creatine kinase-1, CK; peripheral benzodiazipine receptor, PBR; cyclophilin D, CycD) together with 1 μg of green fluorescent protein expression plasmid were transfected into 293T cells. For its high efficiency in apoptosis induction, only ANT-1 was used with 1 and 4 μg of a luciferase construct. Apoptosis was quantified by staining with propidium iodide and fluorescence-activated cell-sorting analysis 18 h after transfection. The data were normalized to the transfection efficiency as assessed by green fluorescent protein fluorescence. The means and SDs of three independent experiments are shown. B, mapping of the apoptosis-inducing domains in ANT-1 by swap mutants of ANT-1 and ANT-2. One μg of different swap mutants that exchange progressively longer sequences beginning at the NH2 terminus of ANT-1 with the corresponding sequences of ANT-2 were generated and transfected into 293T cells in 6-well plates. A schematic drawing shows the ANT-2 sequences in light shading and the ANT-1 domains in dark shading. The position of the last residue of ANT-2 sequence in the exchange (Ex) mutants are given. One μg of an expression plasmid that allowed us to normalize the apoptosis rate to the transfection efficiency was cotransfected. Apoptosis was detected 18 h after transfection by propidium iodide staining and quantified by fluorescence-activated cell-sorting analysis. The means and SDs of three independent transfections for each construct are shown. C, independence of the ANT-1 apoptosis induction from an intact respiratory chain. HeLa ρ0 cells that are deficient in the respiratory chain were generated by prolonged incubation in ethidium bromide. The cells were then investigated for their dependence on addition of uridine to the medium, which is a marker for the absence of mitochondrial DNA (top panels). Scale bar, 100 μm. The presence of the mitochondrially encoded cytochrome oxidase subunit 1 (COX I) was investigated in a RNA blot of HeLa ρ0 cells, and a control against β-actin showed equal loading of the lanes (middle panels). Comparison of ANT-1- and RIP-induced apoptosis in wild-type HeLa and HeLa ρ0 cells is shown (bottom panels). Cells were transfected with the indicated expression plasmids (0.2 μg of ANT-1 or RIP plasmid with 50 ng of green fluorescent protein expression plasmid), and apoptosis was quantified by a fluorescence-activated cell-sorting analysis after 30 h.

The respiratory chain is supposed to be responsible for the generation of reactive oxygen species that are frequently formed during apoptosis and promote cell death induction (27) . Unrestricted reactive oxygen intermediates formation in mitochondria can cause dilated cardiomyopathy (28) , a disease in which ANT-1 is strongly up-regulated (19) . We therefore wanted to determine the contribution of the respiratory chain to ANT-1-induced apoptosis. We created HeLa ρ0 cells that are deficient in the respiratory chain by incubating the cells with low concentrations of ethidium bromide for a prolonged period of time. Fig. 1C ⇓ shows that these cells are dependent on uridine addition, which is indicative of cells with an inactive respiratory chain (21) . In addition, we observed that cytochrome oxidase that is encoded by the mitochondrial DNA is absent in these cells. In the transfections we used conditions that resulted in a relatively weak extent of cell death to be able to assess even slight differences in apoptosis induction. The bottom panel of Fig. 1C ⇓ reveals that ANT-1 could induce apoptosis as potently in HeLa ρ0 cells as in wild-type HeLa cells. RIP, a death domain-containing protein kinase (22) , was used as a control apoptosis inducer and was likewise equally efficient for cell death induction in these two cell lines. However, tumor necrosis factor (TNF) induced a much weaker apoptosis in HeLa ρ0 cells. In addition, cytochrome bL, a component of complex II of the respiratory chain that was isolated as an apoptosis inducer, was inactive in these cells (29) .

Cyclophilin D Specifically Reduces ANT-1-Induced Apoptosis.

The above result, together with the mapping experiment, suggested that specific protein-protein interactions, not the transport activity of ANT-1, are responsible for apoptosis induction. A large body of work on apoptosis suggests that it is mediated by specific protein-protein interactions. Consequently, both repressors and inducers exhibit their function on overexpression. We were therefore interested whether other components of the PT-pore that we have shown to be inactive for apoptosis induction (Fig. 1A) ⇓ could influence ANT-1-induced apoptosis when coexpressed with ANT-1. Fig. 2A ⇓ reveals that only creatine kinase and cyclophilin D could significantly reduce cell death. Because cyclophilin D was the strongest repressor, we focused on this gene for further analysis. We also observed a repression of ANT-1-induced apoptosis in HeLa cells by cyclophilin D (data not shown). Because both proteins, cyclophilin D and ANT-1, are located in mitochondria, we wanted to address the question of whether cotransfected cyclophilin D reduces the import of ANT-1 and therefore leads to a reduction of apoptosis. Fig. 2B ⇓ shows that even a slightly increased amount of ANT-1 protein could be found in mitochondrial extracts when cyclophilin D was cotransfected.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Investigation of the apoptosis repression effect exerted by cyclophilin D. A, apoptosis inhibition activity of the various components of the permeability transition-pore for adenine nucleotide translocator-1 (ANT-1) apoptosis. A luciferase vector (Luc) or the indicated components of the permeability transition (adenine nucleotide translocator-2, ANT-2; voltage-dependent anion channel-1, VDAC I; creatine kinase-1, CK; peripheral benzodiazipine receptor, PBR; cyclophilin D, CycD) were cotransfected in a 3-fold excess over an expression plasmid for ANT-1 (1 μg) in 6-well plates. One μg of a green fluorescence protein expression plasmid was cotransfected to normalize cell death to transfection efficiency. After 18 h, apoptosis was quantified by propidium iodide staining and fluorescence-activated cell-sorting analysis. The means and SDs of three independent measurements are given. B, cyclophilin D cotransfection does not inhibit the import of ANT-1 into mitochondria. 293T cells were transfected in 10-cm plates with expression plasmids for luciferase or for ANT-1 or ANT-1 and cyclophilin D (at a ratio of 1:1 or 1:5). The luciferase plasmid was used to adjust the amounts of transfected DNA. Twenty-four h later, mitochondrial extracts were prepared and probed in a Western blot with the indicated antibodies against the myc epitope or cyclophilin D. The mitochondrial protein Tim23 was used as a loading control for the mitochondrial extracts. C, cyclophilin D repression is independent of its activity as a peptidylprolyl isomerase. An expression construct for ANT-1 (1 μg) together with a 3-fold excess of wild-type cyclophilin or a mutated cyclophilin D (CyloD*R97A/H168Q) that destroyed its catalytic center as a peptidylprolyl isomerase were cotransfected. One μg of an expression plasmid for green fluorescent protein was also used to normalize apoptosis to transfection efficiency. Apoptosis was quantified by fluorescence-activated cell-sorting analysis 18 h later. The means and SDs of three independent measurements are given. D, other mitochondrial chaperones such as p60 and its cochaperone p10 do not repress apoptosis induced by ANT-1. ANT-1 was cotransfected with expression vectors for luciferase, cyclophilin D, or p10 and p60 at a ratio of 1:3. Apoptosis was determined after 28 h using fluorescence-activated cell-sorting analysis. Shown are the means and SDs of three independent experiments. E, cyclophilin D inhibits the breakdown of the mitochondrial membrane potential (ΔΨ) after Ca2+ influx. 293T cells were transfected with the indicated expression plasmids. After 36 h, the cells were incubated with the Ca2+-specific ionophore ionomycin (2.5 μm) and 3,3′-dihexyloxacarbocyanine iodide (0.5 nm). The fluorescence of 3,3′-dihexyloxacarbocyanine iodide was measured by fluorescence-activated cell-sorting analysis at the indicated time points.

Based on the sequence homology, cyclophilin D is a member of the cyclophilin family of chaperones, some of which have been found to regulate apoptosis, such as Hsp70 (30) and Hsp90 (31) . In fact, cyclophilin D has been shown to possess peptidylprolyl isomerase activity (32) . Hence, we were interested in determining whether the peptidylprolyl isomerase activity of cyclophilin D is important for apoptosis reduction. A construct with two point mutations near and in the hydrophobic binding pocket that have both been shown to abrogate the peptidylprolyl isomerase activity in cyclophilins (33) was generated. As Fig. 2C ⇓ demonstrates, this construct could still reduce ANT-1-induced apoptosis. In addition, we observed that the cotransfection of two other mitochondrial chaperones, p10 and p60, which together form an active protein folding complex in mitochondria (34) , were inactive for apoptosis repression (Fig. 2D) ⇓ . These results indicate that the two activities of cyclophilin D, apoptosis regulation and protein folding, can be functionally separated.

Common to pathways that stimulate the PT-pore for apoptosis induction is the breakdown of membrane potential (ΔΨ) over the inner mitochondrial membrane (35) . We therefore tested this membrane potential using 3,3′-dihexyloxacarbocyanine iodide, which accumulates in mitochondria with an intact membrane gradient. The Ca2+-specific ionophore ionomycin served as a PT-pore inducer. It is known that Ca2+ influx activates the PT-pore (3) . Fig. 2E ⇓ shows that on cyclophilin D expression, the number of cells with a reduced mitochondrial membrane potential was decreased.

Apoptosis Repression by Cyclophilin D Correlates with Cell Death Reduction by Bongkrekic Acid (BA), a Specific PT-Pore Inhibitor.

To assess the specificity of cyclophilin D for the repression of ANT-1-induced cell death, we have cotransfected this gene with a number of proapoptotic genes and compared apoptosis repression by cyclophilin D with cell death inhibition by BA. BA is a compound that specifically associates with ANT-1 and inhibits the activation of the PT-pore (36) . As a further control, we used Bcl-2 and Bcl-XL, two known inhibitors of the Bcl-2 gene family. First, we tested two proapoptotic components of the PT-pore for their repression profile. Fig. 3, A and B ⇓ , shows that whereas ANT-1-induced apoptosis could efficiently be repressed by cyclophilin D, cell death by Bax expression was only moderately inhibited by cotransfected cyclophilin D. Likewise, BA could potently diminish apoptosis induced by ANT-1 but was only a weak inhibitor for Bax (Fig. 3C) ⇓ . Of note, Bcl-XL behaved differently with these two apoptosis inducers: it was a strong repressor of Bax-induced apoptosis but was inactive for ANT-1-mediated cell death. Subsequently, we tested apoptosis inducers that are not components of the PT-pore and might therefore activate this protein complex in a more indirect way. Caspase-2 is a caspase that can be found in the nucleus and the Golgi complex (37) and is activated by a diverse range of signals (38) . Recently, it was shown that this protease mediates cytotoxic stress directly rather than only amplifying a mitochondrial cell death signal (39) . Overexpressed caspase-2 was not inhibitable by both cotransfected cyclophilin D and BA (Fig. 4A) ⇓ . Caspase-8 can be found in the active TNF- and the Fas-receptor complex (40) but in addition seems to initiate a proapoptotic signal at the endoplasmic reticulum (41) . Cell death by this protease could be repressed by about 50% by both cotransfected cyclophilin D and BA treatment (Fig. 4B) ⇓ . RIP, another component of the TNF receptor complex, could only be partially inhibited by cyclophilin D. This is mirrored by a moderate reduction of RIP-mediated cell death after application of BA (Fig. 4C) ⇓ . Consequently, BA and cyclophilin D correlate in their activity to diminish apoptosis induced by both components of the PT-pore and apoptosis inducers residing at other locations in the cell.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Cyclophilin D repression of apoptosis by direct activators of the permeability transition-pore correlates with the activity of bongkrekic acid (BA), a specific inhibitor of the permeability transition-pore. A, repression profile of Bax. One μg of expression plasmid for Bax, 0.5 μg of enhanced green fluorescent protein, and 10 μg of an expression plasmid for cyclophilin D, Bcl-XL, or Bcl-2 were cotransfected. Eighteen h later, apoptosis was quantified by fluorescence-activated cell-sorting analysis. B, repression profile of adenine nucleotide translocator-1 (ANT-1). The experimental conditions were the same as those described in A. C, effect of BA on Bax- and ANT-1-induced apoptosis. 293T cells were transfected in 24-well plates with 0.3 μg of expression plasmid for Bax or ANT-1 together with 0.5 μg of enhanced green fluorescent protein. Five h after the transfection, half of the samples were treated with BA at a concentration of 65 μm. Sixteen h later, the cells were analyzed by propidium iodide staining and fluorescence-activated cell-sorting analysis.

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Cyclophilin D repression of general inducers of apoptosis correlates with the activity of bongkrekic acid (BA). A, effect of cyclophilin D and BA on cell death induced by caspase-2. One μg of expression plasmid for caspase-2, 1 μg of enhanced green fluorescent protein, and 5 μg of an expression plasmid for cyclophilin D, Bcl-XL, or Bcl-2 were cotransfected. Eighteen h later, apoptosis was quantified by a fluorescence-activated cell-sorting analysis (left panel). 293T cells were transfected in 6-well plates with 1 μg of expression plasmid for caspase-2 or 2 μg of a control luciferase vector together with 1 μg of enhanced green fluorescent protein. Five h after the transfection, half of the samples were treated with BA at a concentration of 65 μm. Sixteen h later, the cells were analyzed by propidium iodide staining and fluorescence-activated cell-sorting analysis (right panel). B, effect of cyclophilin D and BA on caspase-8-induced apoptosis. The experimental conditions were the same as those described in A. C, effect of cyclophilin D and BA on RIP-induced apoptosis. The experimental conditions used were the same as those described in A. All figures show the means and SDs of three independent experiments.

Cyclophilin D Inhibits Exogenous Apoptosis Inducers.

The above data suggested that cyclophilin D acts as a specific inhibitor of the PT-pore. Because the PT-pore complex is implicated in many diverse proapoptotic signalings (42) , we wanted to know whether, besides cotransfected genes, cyclophilin D could also reduce cell death by exogenously applied apoptosis stimuli. In contrast to specific genes, these agents can be expected to activate many diverse proapoptotic pathways in the cell. We chose TNF and UV irradiation because they induce apoptosis by different pathways that nevertheless activate the PT-pore (43 , 44) . In addition, we used arsenic trioxide, a therapeutic agent used in the treatment of acute promyelocytic leukemias that, although its mode of action is less known, also seems to activate the PT-pore (45) . We performed these experiments in HeLa cells to assess how general the repression activities of cyclophilin D in other cell types are. Fig. 5 ⇓ reveals that cyclophilin D was also active as an apoptosis repressor in HeLa cells. Both apoptosis inducers, TNF and UV, could be repressed in cyclophilin D-transfected cells, and TNF was more efficiently inhibited. Fig. 5C ⇓ shows that the proapoptotic activity of arsenic trioxide could also be inhibited by cyclophilin D.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Cyclophilin D represses apoptosis induced by exogenous cell death stimuli in HeLa cells. The indicated plasmids (1.5 μg) together with 0.5 μg of an expression plasmid for green fluorescent protein were transfected in 10-cm plates with HeLa cells. After 23 h, cells were induced with UV [25 mJ, A; TNF/cycloheximide (15 ng/ml; 0.1 μg/ml), B; or 10 μm arsenic trioxide, C]. Apoptosis was quantified by fluorescence-activated cell-sorting analysis after 18 h. The means and SDs of three independent experiments are shown, normalized to the transfection efficiency.

Cyclophilin D Is Up-Regulated in Specific Human Tumors.

Because apoptosis is suppressed in almost every tumor to overcome the many adverse conditions tumor cells encounter, cell death repression is supposed to be a hallmark of cancer cells (12) . Consequently, many genes that inhibit proapoptotic stimuli are up-regulated in tumors (46 , 47) . We have found cyclophilin D to repress apoptosis induction via the PT-pore, which is activated by many diverse stimuli. We therefore tested the expression level of cyclophilin D in a tumor expression profiling blot that covers a number of different human tumors. This membrane contains RNA of both normal and tumor tissue from each patient. Fig. 6 ⇓ shows that cyclophilin D is specifically and significantly up-regulated in human tumors of the breast, uterus, and ovary. The other tested tumors displayed a normal distribution of the expression level of this gene.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Cyclophilin D is specifically up-regulated in human tumors of the breast, uterus, and ovary. A multiple tumor dot-blot was hybridized with human cyclophilin D probe. The number of patients is given below the tissue that was investigated. The signal intensities were measured with densitometry, and the ln-values of the ratios between tumor and normal signals were summed for each patient and plotted. Down- or up-regulation by a factor of 1.5 (ln1.5 = 0.4) was regarded as significant and is indicated by the gray-shaded areas.

DISCUSSION

Because the regulation of apoptosis is of profound relevance for a number of diseases, especially cancer, its molecular regulation is the focus of intense investigations. In this report we have further analyzed apoptosis induction by ANT-1, a component of the PT-pore that we found among genes from a screen for proapoptotic genes (16 , 17 , 29 , 48 , 49) . Importantly, ANT-1 is up-regulated in the prevalent degenerative heart disease dilated cardiomyopathy (19) , which is characterized by an excess of apoptotic myocytes early in the progression of this fatal disease (20) . A multitude of data suggest that apoptosis is mediated by specific protein-protein interactions. Consequently, both repressors and inducers exhibit their activity on overexpression. Using this correlation, our molecular analysis of ANT-1-induced apoptosis led us to its interactor, cyclophilin D, which is able to repress ANT-1-induced cell death. Additional studies indicated that cyclophilin D inhibited the activation of the PT-pore by many other proapoptotic stimuli. In this study we have also demonstrated that cyclophilin D is significantly up-regulated in various tumors of reproductive tissues (Fig. 6) ⇓ . Consequently, starting with a gene involved in a degenerative heart disease (ANT-1), our studies proceeded to a gene that seems to be involved in a proliferative disease (cyclophilin D). We have therefore collected evidence that the PT-pore contains at least two proteins relevant for diseases.

We propose that cyclophilin D represents a new kind of apoptosis inhibitor that is effective at a functionally different level than the known inhibitors of the Bcl-2 family. First, the antiapoptotic activity of cyclophilin D does not correlate with the established cell death inhibitors Bcl-2 and Bcl-XL (Fig. 3, B and C) ⇓ . Furthermore, whereas the Bcl-2 proteins can also function independently of the PT-pore (7) , cyclophilin D appears to be specific for this protein complex (see below). Lastly, cyclophilin D interacts with ANT-1, a component of the mitochondrial inner membrane, and is therefore also locally separated from the Bcl-2 system. Given the involvement of ANT-1 in dilated cardiomyopathy and cyclophilin D in tumors, our findings could therefore have a therapeutic value.

We suggest that cyclophilin D is specific for the PT-pore in its activity to repress apoptosis. Several lines of evidence support this view. First, cyclophilin D displays the same cell death repression profile as BA, which is a known specific inhibitor of the PT-pore (Figs. 3 ⇓ and 4 ⇓ ). Secondly, cyclophilin D is a potent repressor of ANT-1-induced apoptosis, which seems to activate the PT-pore directly for apoptosis induction (17) . Thirdly, cyclophilin D is a component of the PT-pore. To our knowledge, it has not been found in other protein complexes in the cell. It is therefore likely that cyclophilin D is confined in its activity to the PT-pore. In line with this, a cyclophilin D construct devoid of its mitochondrial import signal showed a dramatic reduction of apoptosis repression (data not shown). Furthermore, the cell death repression is independent of its peptidyl prolylisomerase activity (Fig. 2C) ⇓ . It is therefore likely that permanent interactions, such as those with components of the PT-pore [especially the strong association with ANT-1 (50)] , and not transient protein contacts, are relevant for its antiapoptotic function. How would the interactions that allow cyclophilin D to act as a repressor for apoptosis induction appear? The PT-pore is converted into a nonspecific channel on apoptosis induction (42) . Consequently, cyclophilin D could act to close the PT-pore channel and only allows a controlled exchange of ADP and ATP. This model would require that cyclophilin D is inactivated during cell death. In fact, a recent paper noted that cyclophilin D mRNA is degraded on apoptosis induction (51) . This could be one mechanism (but not necessary the only one) for the inactivation of cyclophilin D. Other, additional mechanisms could be conformational changes or the recruitment of additional binding partners.

In former models, cyclophilin D has been supposed to act in a proapoptotic way because cyclosporin that binds and inhibits cyclophilins can also inhibit PT-pore activation and apoptosis induction (42 , 52 , 53) . However, because the cell contains at least eight different cyclophilins outside of the mitochondria, the specificity of the cyclosporin effect on the PT-pore has been questioned (54) . In line with this, we observed that cyclosporin A could reduce ANT-1-induced apoptosis in 293T cells by only 20% under the best conditions (data not shown), suggesting that this compound acts differently from cyclophilin D. In addition, there is accumulating evidence that demonstrates that cyclosporin can also induce apoptosis (55, 56, 57) . Furthermore, the inhibitory function of cyclosporins is based on the ability of cyclophilins to act as isomerases (52 , 53) . Because we have found that the antiapoptotic function of cyclophilin D is not dependent on its peptidylprolyl isomerase activity activity, other effects must be responsible for the inhibitory or inducing effects of cyclosporin A. In contrast to our study, a recent report showed that cyclophilin D can function as a repressor for apoptosis only when it is functional as a peptidylprolyl isomerase (58) . However, in these experiments, only the potential of the inner membrane of mitochondria was taken as an indicator for apoptosis, which is fraught with potential artifacts (1) and does not always correlate with cell death induction (7 , 59) . When apoptosis was measured by membrane permeability, these authors observed a temporal delay of only 2 h by cyclophilin D expression. We obtained much stronger effects using the degradation of genomic DNA as an established parameter for apoptosis. Our finding that cyclophilin D has an activity independent of its peptidylprolyl isomerase activity (Fig. 2C) ⇓ is not without precedent: cyclophilin A can mediate HIV replication by interacting with Gag without its isomerase function (60) , and the porcine kidney peptidylprolyl isomerase can fold the creatine kinase without its catalytic activity (61) .

Given the possible involvement of ANT-1 in the heart disease dilated cardiomyopathy (17 , 19) , we investigated apoptosis by ANT-1. Although this ADP/ATP transporter can be regarded as an indirect functional component of the respiratory chain, this protein complex seems to be dispensable for cell death (Fig. 1C) ⇓ . Our results also show that the PT-pore components ANT-1 and Bax activate cell death differently (5 , 6) because the antiapoptotic Bcl-XL could distinguish between these apoptosis inducers (Fig. 3) ⇓ . Furthermore, BA and cyclophilin D have differing effectiveness in reducing the effects of these proapoptotic genes (Fig. 3) ⇓ . We also found that specific sequences are required for ANT-1 to induce apoptosis (Fig. 1B) ⇓ . According to a proposed structure of ANT-1 (26) , the domains responsible for apoptosis induction lie on both sides of the inner mitochondrial membrane: whereas the third loop protrudes into the matrix; the NH2 terminus and the second and fourth loop face the intermembrane space. Consequently, assuming that cyclophilin D resides in the matrix of mitochondria, only the third loop that faces this compartment could be bound and inhibited by cyclophilin D. However, cyclophilin D has also been found in the innermembrane space and could therefore contribute to the repression of these apoptosis-inducing domains of ANT-1 (32) . An additional putative inhibitor of ANT-1 is the creatine kinase that is also located in the innermembrane space. Of note, this molecule could likewise, but less efficiently, reduce ANT-1-mediated cell death (Fig. 2A) ⇓ , and its ligand, creatine, reduces apoptosis in many different diseases ranging from Alzheimer’s disease to amyotrophic lateral sclerosis (62, 63, 64) . This underscores the validity of our approach with cotransfected PT-pore components. Because the PT-pore is supposed to consist of additional proteins (4) , other, previously unknown constituents could also act as inhibitors of ANT-1.

We have found that cyclophilin D is significantly increased in tumors of the breast, uterus, and ovary (Fig. 6) ⇓ . If the PT-pore is involved in so many kinds of apoptosis induction by being seemingly activated by a multitude of different apoptosis inducers (42) , why is cyclophilin D only up-regulated in tumors associated with reproductive tissues in females? Whereas a definitive answer cannot be given at this time, these tissues are estrogen dependent. It is known that apoptosis repression is especially important for tumorigenesis of such tissues (65 , 66) . There is also a growing body of evidence that cyclophilins play a role in the development of tumors (67, 68, 69) . Consequently, it is possible that cyclophilin D is involved in apoptosis repression and hence in the growth and progression of hormone-dependent tumors.

Expression of proapoptotic genes allows induction of specific signals in the cell for apoptosis induction. Not all components of the PT-pore are active for apoptosis induction or repression (Figs. 1A ⇓ and 2A ⇓ ). This stresses the specificity of the protein-protein contacts that are responsible for cell death induction. In addition, this could also indicate that some of the PT-pore components might not be involved in apoptosis regulation. After all, the PT-pore has the important function in healthy cells of transporting metabolic intermediates in and out of the mitochondria. Whereas the role of the PT-pore itself in apoptosis induction has firmly been established, the contribution of the various components has been less clear. We have demonstrated here that the PT-pore contains both repressors and inducers of apoptosis, both of which are relevant for specific diseases.

Acknowledgments

We thank F. Voss for excellent technical assistance and C. Dalibor for technical support.

Footnotes

  • Grant support: The Bavarian Government, Roche Diagnostics, and Xantos Biomedicine AG.

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

  • Requests for reprints: Stefan Grimm, Max-Planck-Institute for Biochemistry, Am Klopferspitz 18a, 82152 Martinsried, Germany. Phone: 49-89-8578-2219; Fax: 49-89-8578-3888; E-mail: sgrimm{at}biochem.mpg.de

  • Received March 1, 2003.
  • Revision received October 6, 2003.
  • Accepted October 28, 2003.
  • ©2004 American Association for Cancer Research.

References

  1. ↵
    Green D. R., Reed J. C. Mitochondria and apoptosis. Science (Wash. DC), 281: 1309-1312, 1998.
    OpenUrlAbstract/FREE Full Text
  2. ↵
    Halestrap A. P. The mitochondrial permeability transition: its molecular mechanism and role in reperfusion injury. Biochem. Soc. Symp., 66: 181-203, 1999.
    OpenUrlPubMed
  3. ↵
    Crompton M. The mitochondrial permeability transition pore and its role in cell death. Biochem. J., 341: 233-249, 1999.
  4. ↵
    Marzo I., Brenner C., Zamzami N., Susin S. A., Beutner G., Brdiczka D., Remy R., Xie Z. H., Reed J. C., Kroemer G. The permeability transition pore complex: a target for apoptosis regulation by caspases and bcl-2-related proteins. J. Exp. Med., 187: 1261-1271, 1998.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Shimizu S., Narita M., Tsujimoto Y. Bcl-2 family proteins regulate the release of apoptogenic cytochrome c by the mitochondrial channel VDAC. Nature (Lond.), 399: 483-487, 1999.
    OpenUrlCrossRefPubMed
  6. ↵
    Marzo I., Brenner C., Zamzami N., Jurgensmeier J. M., Susin S. A., Vieira H. L., Prevost M. C., Xie Z., Matsuyama S., Reed J. C., Kroemer G. Bax and adenine nucleotide translocator cooperate in the mitochondrial control of apoptosis. Science (Wash. DC), 281: 2027-2031, 1998.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Eskes R., Antonsson B., Osen-Sand A., Montessuit S., Richter C., Sadoul R., Mazzei G., Nichols A., Martinou J. C. Bax-induced cytochrome c release from mitochondria is independent of the permeability transition pore but highly dependent on Mg2+ ions. J. Cell Biol., 143: 217-224, 1998.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Susin S. A., Lorenzo H. K., Zamzami N., Marzo I., Snow B. E., Brothers G. M., Mangion J., Jacotot E., Costantini P., Loeffler M., Larochette N., Goodlett D. R., Aebersold R., Siderovski D. P., Penninger J. M., Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature (Lond.), 397: 441-446, 1999.
    OpenUrlCrossRefPubMed
  9. ↵
    Verhagen A. M., Ekert P. G., Pakusch M., Silke J., Connolly L. M., Reid G. E., Moritz R. L., Simpson R. J., Vaux D. L. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell, 102: 43-53, 2000.
    OpenUrlCrossRefPubMed
  10. ↵
    Du C., Fang M., Li Y., Li L., Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell, 102: 33-42, 2000.
    OpenUrlCrossRefPubMed
  11. ↵
    Liu X., Kim C. N., Yang J., Jemmerson R., Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell, 86: 147-157, 1996.
    OpenUrlCrossRefPubMed
  12. ↵
    Hanahan D., Weinberg R. A. The hallmarks of cancer. Cell, 100: 57-70, 2000.
    OpenUrlCrossRefPubMed
  13. ↵
    Costantini P., Jacotot E., Decaudin D., Kroemer G. Mitochondrion as a novel target of anticancer chemotherapy. J. Natl. Cancer Inst. (Bethesda), 92: 1042-1053, 2000.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Rampino N., Yamamoto H., Ionov Y., Li Y., Sawai H., Reed J. C., Perucho M. Somatic frameshift mutations in the BAX gene in colon cancers of the microsatellite mutator phenotype. Science (Wash. DC), 275: 967-969, 1997.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Vaux D. L., Cory S., Adams J. M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature (Lond.), 335: 440-442, 1988.
    OpenUrlCrossRefPubMed
  16. ↵
    Grimm S., Leder P. An apoptosis-inducing isoform of neu differentiation factor (NDF) identified using a novel screen for dominant, apoptosis-inducing genes. J. Exp. Med., 185: 1137-1142, 1997.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Bauer M. K. A., Schubert A., Rocks O., Grimm S. Adenine nucleotide translocase-1, a component of the permeability transition pore, can dominantly induce apoptosis. J. Cell Biol., 147: 1493-1502, 1999.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Belzacq A. S., El Hamel C., Vieira H. L., Cohen I., Haouzi D., Metivier D., Marchetti P., Brenner C., Kroemer G. Adenine nucleotide translocator mediates the mitochondrial membrane permeabilization induced by lonidamine, arsenite and CD437. Oncogene, 20: 7579-7587, 2001.
    OpenUrlCrossRefPubMed
  19. ↵
    Dorner A., Schulze K., Rauch U., Schultheiss H. P. Adenine nucleotide translocator in dilated cardiomyopathy: pathophysiological alterations in expression and function. Mol. Cell. Biochem., 174: 261-269, 1997.
    OpenUrlCrossRefPubMed
  20. ↵
    Narula J., Kolodgie F. D., Virmani R. Apoptosis and cardiomyopathy. Curr. Opin. Cardiol., 15: 183-188, 2000.
    OpenUrlCrossRefPubMed
  21. ↵
    King M. P., Attardi G. Human cells lacking mtDNA: repopulation with exogenous mitochondria by complementation. Science (Wash. DC), 246: 500-503, 1989.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Stanger B. Z., Leder P., Lee T. H., Kim E., Seed B. RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell, 81: 513-523, 1995.
    OpenUrlCrossRefPubMed
  23. ↵
    Bitzer M., Prinz F., Bauer M., Spiegel M., Neubert W. J., Gregor M., Schulze-Osthoff K., Lauer U. Sendai virus infection induces apoptosis through activation of caspase-8 (FLICE) and caspase-3 (CPP32). J. Virol., 73: 702-708, 1999.
    OpenUrlAbstract/FREE Full Text
  24. ↵
    Belzacq A. S., Vieira H. L., Verrier F., Vandecasteele G., Cohen I., Prevost M. C., Larquet E., Pariselli F., Petit P. X., Kahn A., Rizzuto R., Brenner C., Kroemer G. Bcl-2 and Bax modulate adenine nucleotide translocase activity. Cancer Res., 63: 541-546, 2003.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Manfredi G., Yang L., Gajewski C. D., Mattiazzi M. Measurements of ATP in mammalian cells. Methods, 26: 317-326, 2002.
    OpenUrlCrossRefPubMed
  26. ↵
    Klingenberg M., Nelson D. R. Structure-function relationships of the ADP/ATP carrier. Biochim. Biophys. Acta, 1187: 241-244, 1994.
    OpenUrlPubMed
  27. ↵
    Simon H. U., Haj-Yehia A., Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis, 5: 415-418, 2000.
    OpenUrlCrossRefPubMed
  28. ↵
    Li Y., Huang T. T., Carlson E. J., Melov S., Ursell P. C., Olson J. L., Noble L. J., Yoshimura M. P., Berger C., Chan P. H., et al Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat. Genet., 11: 376-381, 1995.
    OpenUrlCrossRefPubMed
  29. ↵
    Albayrak T., Scherhammer V., Schoenfeld N., Braziulis E., Mund T., Bauer M. K., Scheffler I. E., Grimm S. The tumor suppressor cybL, a component of the respiratory chain, mediates apoptosis induction. Mol. Biol. Cell, 14: 3082-3096, 2003.
    OpenUrlAbstract/FREE Full Text
  30. ↵
    Beere H. M., Wolf B. B., Cain K., Mosser D. D., Mahboubi A., Kuwana T., Tailor P., Morimoto R. I., Cohen G. M., Green D. R. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 apoptosome. Nat. Cell Biol., 2: 469-475, 2000.
    OpenUrlCrossRefPubMed
  31. ↵
    Pandey P., Saleh A., Nakazawa A., Kumar S., Srinivasula S. M., Kumar V., Weichselbaum R., Nalin C., Alnemri E. S., Kufe D., Kharbanda S. Negative regulation of cytochrome c-mediated oligomerization of Apaf-1 and activation of procaspase-9 by heat shock protein 90. EMBO J., 19: 4310-4322, 2000.
    OpenUrlAbstract
  32. ↵
    Tanveer A., Virji S., Andreeva L., Totty N. F., Hsuan J. J., Ward J. M., Crompton M. Involvement of cyclophilin D in the activation of a mitochondrial pore by Ca2+ and oxidant stress. Eur. J. Biochem., 238: 166-172, 1996.
    OpenUrlPubMed
  33. ↵
    Zydowsky L. D., Etzkorn F. A., Chang H. Y., Ferguson S. B., Stolz L. A., Ho S. I., Walsh C. T. Active site mutants of human cyclophilin A separate peptidyl-prolyl isomerase activity from cyclosporin A binding and calcineurin inhibition. Protein Sci., 1: 1092-1099, 1992.
    OpenUrlCrossRefPubMed
  34. ↵
    Ryan M. T., Naylor D. J., Hoj P. B., Clark M. S., Hoogenraad N. J. The role of molecular chaperones in mitochondrial protein import and folding. Int. Rev. Cytol., 174: 127-193, 1997.
    OpenUrlCrossRefPubMed
  35. ↵
    Zamzami N., Brenner C., Marzo I., Susin S. A., Kroemer G. Subcellular and submitochondrial mode of action of Bcl-2-like oncoproteins. Oncogene, 16: 2265-2282, 1998.
    OpenUrlCrossRefPubMed
  36. ↵
    Klingenberg M., Grebe K., Heldt H. W. On the inhibition of the adenine nucleotide translocation by bongkrekic acid. Biochem. Biophys. Res. Commun., 39: 344-351, 1970.
    OpenUrlCrossRefPubMed
  37. ↵
    Mancini M., Machamer C. E., Roy S., Nicholson D. W., Thornberry N. A., Casciola-Rosen L. A., Rosen A. Caspase-2 is localized at the Golgi complex and cleaves golgin-160 during apoptosis. J. Cell Biol., 149: 603-612, 2000.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Harvey N. L., Butt A. J., Kumar S. Functional activation of Nedd2/ICH-1 (caspase-2) is an early process in apoptosis. J. Biol. Chem., 272: 13134-13139, 1997.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Lassus P., Opitz-Araya X., Lazebnik Y. Requirement for caspase-2 in stress-induced apoptosis before mitochondrial permeabilization. Science (Wash. DC), 297: 1352-1354, 2002.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Ashkenazi A., Dixit V. M. Apoptosis control by death and decoy receptors. Curr. Opin. Cell Biol., 11: 255-260, 1999.
    OpenUrlCrossRefPubMed
  41. ↵
    Ng F. W., Nguyen M., Kwan T., Branton P. E., Nicholson D. W., Cromlish J. A., Shore G. C. p28 Bap31, a Bcl-2/Bcl-XL- and procaspase-8-associated protein in the endoplasmic reticulum. J. Cell Biol., 139: 327-338, 1997.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Kroemer G., Zamzami N., Susin S. A. Mitochondrial control of apoptosis. Immunol. Today, 18: 44-51, 1997.
    OpenUrlCrossRefPubMed
  43. ↵
    Scorrano L., Penzo D., Petronilli V., Pagano F., Bernardi P. Arachidonic acid causes cell death through the mitochondrial permeability transition. Implications for tumor necrosis factor-α apoptotic signaling. J. Biol. Chem., 276: 12035-12040, 2001.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    Stoebner P. E., Carayon P., Casellas P., Portier M., Lavabre-Bertrand T., Cuq P., Cano J. P., Meynadier J., Meunier L. Transient protection by peripheral benzodiazepine receptors during the early events of ultraviolet light-induced apoptosis. Cell Death Differ., 8: 747-753, 2001.
    OpenUrlCrossRefPubMed
  45. ↵
    Larochette N., Decaudin D., Jacotot E., Brenner C., Marzo I., Susin S. A., Zamzami N., Xie Z., Reed J., Kroemer G. Arsenite induces apoptosis via a direct effect on the mitochondrial permeability transition pore. Exp. Cell Res., 249: 413-421, 1999.
    OpenUrlCrossRefPubMed
  46. ↵
    Baliga B. C., Kumar S. Role of Bcl-2 family of proteins in malignancy. Hematol. Oncol., 20: 63-74, 2002.
    OpenUrlCrossRefPubMed
  47. ↵
    LaCasse E. C., Baird S., Korneluk R. G., MacKenzie A. E. The inhibitors of apoptosis (IAPs) and their emerging role in cancer. Oncogene, 17: 3247-3259, 1998.
    OpenUrlCrossRefPubMed
  48. ↵
    Mund T., Gewies A., Schoenfeld N., Bauer M. K., Grimm S. Spike, a novel BH3-only protein, regulates apoptosis at the endoplasmic reticulum. FASEB J., 17: 696-698, 2003.
    OpenUrlAbstract/FREE Full Text
  49. ↵
    Schoenfeld, N., Bauer, M. K. A., and Grimm, S. The metastasis suppressor gene C33/CD82/KAI1 induces apoptosis through reactive oxygen intermediates. FASEB J., in press, 2003.
  50. ↵
    Crompton M., Virji S., Ward J. M. Cyclophilin-D binds strongly to complexes of the voltage-dependent anion channel and the adenine nucleotide translocase to form the permeability transition pore. Eur. J. Biochem., 258: 729-735, 1998.
    OpenUrlCrossRefPubMed
  51. ↵
    Hursting S. D., Shen J. C., Sun X. Y., Wang T. T., Phang J. M., Perkins S. N. Modulation of cyclophilin gene expression by N-4-(hydroxyphenyl)retinamide: association with reactive oxygen species generation and apoptosis. Mol. Carcinog., 33: 16-24, 2002.
    OpenUrlCrossRefPubMed
  52. ↵
    Halestrap A. P., Davidson A. M. Inhibition of Ca2+-induced large-amplitude swelling of liver and heart mitochondria by cyclosporin is probably caused by the inhibitor binding to mitochondrial-matrix peptidyl-prolyl cis-trans isomerase and preventing it interacting with the adenine nucleotide translocase. Biochem. J., 268: 153-160, 1990.
    OpenUrlAbstract/FREE Full Text
  53. ↵
    McGuinness O., Yafei N., Costi A., Crompton M. The presence of two classes of high-affinity cyclosporin A binding sites in mitochondria. Evidence that the minor component is involved in the opening of an inner-membrane Ca2+-dependent pore. Eur. J. Biochem., 194: 671-679, 1990.
    OpenUrlPubMed
  54. ↵
    Martinou J. C., Green D. R. Breaking the mitochondrial barrier. Nat. Rev. Mol. Cell Biol., 2: 63-67, 2001.
    OpenUrlCrossRefPubMed
  55. ↵
    Esposito C., Fornoni A., Cornacchia F., Bellotti N., Fasoli G., Foschi A., Mazzucchelli I., Mazzullo T., Semeraro L., Dal Canton A. Cyclosporine induces different responses in human epithelial, endothelial and fibroblast cell cultures. Kidney Int., 58: 123-130, 2000.
    OpenUrlCrossRefPubMed
  56. ↵
    Gottschalk A. R., Boise L. H., Thompson C. B., Quintans J. Identification of immunosuppressant-induced apoptosis in a murine B-cell line and its prevention by bcl-x but not bcl-2. Proc. Natl. Acad. Sci. USA, 91: 7350-7354, 1994.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    Damoiseaux J. G., Defresne M. P., Reutelingsperger C. P., Van Breda Vriesman P. J. Cyclosporin-A differentially affects apoptosis during in vivo rat thymocyte maturation. Scand. J. Immunol., 56: 353-360, 2002.
    OpenUrlCrossRefPubMed
  58. ↵
    Lin D. T., Lechleiter J. D. Mitochondrial targeted cyclophilin D protects cells from cell death by peptidyl prolyl isomerization. J. Biol. Chem., 277: 31134-31141, 2002.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    Finucane D. M., Waterhouse N. J., Amarante-Mendes G. P., Cotter T. G., Green D. R. Collapse of the inner mitochondrial transmembrane potential is not required for apoptosis of HL60 cells. Exp. Cell Res., 251: 166-174, 1999.
    OpenUrlCrossRefPubMed
  60. ↵
    Saphire A. C., Bobardt M. D., Gallay P. A. Trans-complementation rescue of cyclophilin A-deficient viruses reveals that the requirement for cyclophilin A in human immunodeficiency virus type 1 replication is independent of its isomerase activity. J. Virol., 76: 2255-2262, 2002.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    Ou W. B., Luo W., Park Y. D., Zhou H. M. Chaperone-like activity of peptidyl-prolyl cis-trans isomerase during creatine kinase refolding. Protein Sci., 10: 2346-2353, 2001.
    OpenUrlCrossRefPubMed
  62. ↵
    Brewer G. J., Wallimann T. W. Protective effect of the energy precursor creatine against toxicity of glutamate and β-amyloid in rat hippocampal neurons. J. Neurochem., 74: 1968-1978, 1968.
    OpenUrlCrossRef
  63. ↵
    Klivenyi P., Ferrante R. J., Matthews R. T., Bogdanov M. B., Klein A. M., Andreassen O. A., Mueller G., Wermer M., Kaddurah-Daouk R., Beal M. F. Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat. Med., 5: 347-350, 1999.
    OpenUrlCrossRefPubMed
  64. ↵
    Malcon C., Kaddurah-Daouk R., Beal M. F. Neuroprotective effects of creatine administration against NMDA and malonate toxicity. Brain Res., 860: 195-198, 2000.
    OpenUrlCrossRefPubMed
  65. ↵
    Lipponen P. Apoptosis in breast cancer: relationship with other pathological parameters. Endocr. Relat. Cancer, 6: 13-16, 1999.
    OpenUrlAbstract
  66. ↵
    Thompson C. B. Apoptosis in the pathogenesis and treatment of disease. Science (Wash. DC), 267: 1456-1462, 1995.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    Kumar P., Mark P. J., Ward B. K., Minchin R. F., Ratajczak T. Estradiol-regulated expression of the immunophilins cyclophilin 40 and FKBP52 in MCF-7 breast cancer cells. Biochem. Biophys. Res. Commun., 284: 219-225, 2001.
    OpenUrlCrossRefPubMed
  68. ↵
    Ryo A., Liou Y. C., Wulf G., Nakamura M., Lee S. W., Lu K. P. PIN1 is an E2F target gene essential for Neu/Ras-induced transformation of mammary epithelial cells. Mol. Cell. Biol., 22: 5281-5295, 2002.
    OpenUrlAbstract/FREE Full Text
  69. ↵
    Kim J. O., Nau M. M., Allikian K. A., Makela T. P., Alitalo K., Johnson B. E., Kelley M. J. Co-amplification of a novel cyclophilin-like gene (PPIE) with L-myc in small cell lung cancer cell lines. Oncogene, 17: 1019-1026, 1998.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 64 (1)
January 2004
Volume 64, Issue 1
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Cyclophilin D, a Component of the Permeability Transition-Pore, Is an Apoptosis Repressor
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Cyclophilin D, a Component of the Permeability Transition-Pore, Is an Apoptosis Repressor
Alexis Schubert and Stefan Grimm
Cancer Res January 1 2004 (64) (1) 85-93; DOI: 10.1158/0008-5472.CAN-03-0476

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Cyclophilin D, a Component of the Permeability Transition-Pore, Is an Apoptosis Repressor
Alexis Schubert and Stefan Grimm
Cancer Res January 1 2004 (64) (1) 85-93; DOI: 10.1158/0008-5472.CAN-03-0476
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • HSF1-Mediated Aneuploidy in p53-Defective Cells
  • Mechanisms of Invasive Urothelial Tumorigenesis
  • Alternate mRNA 3′ Processing Characterizes Distinct Tumors
Show more Molecular Biology, Pathobiology, and Genetics
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement