Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(46): 6490-6500, 2017 11 16.
Article in English | MEDLINE | ID: mdl-28759041

ABSTRACT

The critical role of calcium signalling in processes related to cancer cell proliferation and invasion has seen a focus on pharmacological inhibition of overexpressed ion channels in specific cancer subtypes as a potential therapeutic approach. However, despite the critical role of calcium in cell death pathways, pharmacological activation of overexpressed ion channels has not been extensively evaluated in breast cancer. Here we define the overexpression of transient receptor potential vanilloid 4 (TRPV4) in a subgroup of breast cancers of the basal molecular subtype. We also report that pharmacological activation of TRPV4 with GSK1016790A reduced viability of two basal breast cancer cell lines with pronounced endogenous overexpression of TRPV4, MDA-MB-468 and HCC1569. Pharmacological activation of TRPV4 produced pronounced cell death through two mechanisms: apoptosis and oncosis in MDA-MB-468 cells. Apoptosis was associated with PARP-1 cleavage and oncosis was associated with a rapid decline in intracellular ATP levels, which was a consequence of, rather than the cause of, the intracellular ion increase. TRPV4 activation also resulted in reduced tumour growth in vivo. These studies define a novel therapeutic strategy for breast cancers that overexpress specific calcium permeable plasmalemmal ion channels with available selective pharmacological activators.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , TRPV Cation Channels/genetics , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Immunoblotting , Leucine/analogs & derivatives , Leucine/pharmacology , Mice, Inbred BALB C , Mice, Nude , Necrosis/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism , Xenograft Model Antitumor Assays
2.
Cell Death Differ ; 21(6): 876-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24488096

ABSTRACT

Cytotoxic lymphocytes (CLs) contain lysosome-related organelles (LROs) that perform the normal degradative functions of the lysosome, in addition to storage and release of powerful cytotoxins employed to kill virally infected or abnormal cells. Among these cytotoxins is granzyme B (GrB), a protease that has also been implicated in activation (restimulation)-induced cell death of natural killer (NK) and T cells, but the underlying mechanism and its regulation are unclear. Here we show that restimulation of previously activated human or mouse lymphocytes induces lysosomal membrane permeabilisation (LMP), followed by GrB release from LROs into the CL cytosol. The model lysosomal stressors sphingosine and Leu-Leu-methyl-ester, and CLs from gene-targeted mice were used to show that LMP releases GrB in both a time- and concentration-dependent manner, and that the liberated GrB is responsible for cell death. The endogenous GrB inhibitor Serpinb9 (Sb9) protects CLs against LMP-induced death but is decreasingly effective as the extent of LMP increases. We also used these model stressors to show that GrB is the major effector of LMP-mediated death in T cells, but that in NK cells additional effectors are released, making GrB redundant. We found that limited LMP and GrB release occurs constitutively in proliferating lymphocytes and in NK cells engaged with targets in vitro. In Ectromelia virus-infected lymph nodes, working NK cells lacking Sb9 are more susceptible to GrB-mediated death. Taken together, these data show that a basal level of LMP occurs in proliferating and activated lymphocytes, and is increased on restimulation. LMP releases GrB from LROs into the lymphocyte cytoplasm and its ensuing interaction with Sb9 dictates whether or not the cell survives. The GrB-Sb9 nexus may therefore represent an additional mechanism of limiting lymphocyte lifespan and populations.


Subject(s)
Cell Death/drug effects , Granzymes/metabolism , Serpins/metabolism , Stress, Physiological/genetics , Animals , Cell Membrane Permeability/drug effects , Humans , Killer Cells, Natural/drug effects , Lymphocytes/cytology , Lymphocytes/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Mice , Sphingosine/pharmacology , Stress, Physiological/drug effects , T-Lymphocytes/drug effects
3.
Cell Death Differ ; 20(9): 1183-93, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23744295

ABSTRACT

Human and mouse granzyme (Gzm)B both induce target cell apoptosis in concert with pore-forming perforin (Pfp); however the mechanisms by which other Gzms induce non-apoptotic death remain controversial and poorly characterised. We used timelapse microscopy to document, quantitatively and in real time, the death of target cells exposed to primary natural killer (NK) cells from mice deficient in key Gzms. We found that in the vast majority of cases, NK cells from wild-type mice induced classic apoptosis. However, NK cells from syngeneic Gzm B-deficient mice induced a novel form of cell death characterised by slower kinetics and a pronounced, writhing, 'worm-like' morphology. Dying cells initially contracted but did not undergo membrane blebbing, and annexin-V staining was delayed until the onset of secondary necrosis. As it is different from any cell death process previously reported, we tentatively termed this cell death 'athetosis'. Two independent lines of evidence showed this alternate form of death was due to Gzm A: first, cell death was revealed in the absence of Gzm B, but was completely lost when the NK cells were deficient in both Gzm A and B; second, the athetotic morphology was precisely reproduced when recombinant mouse Gzm A was delivered by an otherwise innocuous dose of recombinant Pfp. Gzm A-mediated athetosis did not require caspase activation, early mitochondrial disruption or generation of reactive oxygen species, but did require an intact actin cytoskeleton and was abolished by latrunculin B and mycalolide B. This work defines an authentic role for mouse Gzm A in granule-induced cell death by cytotoxic lymphocytes.


Subject(s)
Apoptosis/drug effects , Granzymes/metabolism , Killer Cells, Natural/immunology , Perforin/metabolism , Actin Cytoskeleton , Animals , Apoptosis/immunology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Granzymes/deficiency , Granzymes/genetics , HeLa Cells , Humans , Killer Cells, Natural/cytology , Marine Toxins , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Oxazoles/pharmacology , Thiazolidines/pharmacology , Time-Lapse Imaging
4.
Cell Death Dis ; 3: e344, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22764103

ABSTRACT

Overexpression of Bcl-2 contributes to resistance of cancer cells to human cytotoxic lymphocytes (CL) by blocking granzyme B (GraB)-induced mitochondrial outer membrane permeabilization (MOMP). Drugs that neutralise Bcl-2 (e.g., ABT-737) may therefore be effective adjuvants for immunotherapeutic strategies that use CL to kill cancer cells. Consistent with this we found that ABT-737 effectively restored MOMP in Bcl-2 overexpressing cells treated with GraB or natural killer cells. This effect was observed even if ABT-737 was added up to 16 h after GraB, after which the cells reset their resistant phenotype. Sensitivity to ABT-737 required initial cleavage of Bid by GraB (gctBid) but did not require ongoing GraB activity once Bid had been cleaved. This gctBid remained detectable in cells that were sensitive to ABT-737, but Bax and Bak were only activated if ABT-737 was added to the cells. These studies demonstrate that GraB generates a prolonged pro-apoptotic signal that must remain active for ABT-737 to be effective. The duration of this signal is determined by the longevity of gctBid but not activation of Bax or Bak. This defines a therapeutic window in which ABT-737 and CL synergise to cause maximum death of cancer cells that are resistant to either treatment alone, which will be essential in defining optimum treatment regimens.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Granzymes/pharmacology , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Membrane Permeability/drug effects , Cytochromes c/metabolism , HeLa Cells , Humans , Killer Cells, Natural/immunology , Mitochondria/metabolism , Piperazines/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
7.
Cell Death Differ ; 16(4): 537-42, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19136939

ABSTRACT

Time-lapse video microscopy allows analysis of the interaction between individual CTLs and adherent peptide-pulsed targets, from contact, to lymphocyte detachment, APC rounding, phosphatidylserine exposure and finally loss of plasma membrane integrity characteristic of end-stage apoptosis. Using in vitro-stimulated effectors specific for the ovalbumin K(b)OVA(257) (OT-I) and influenza A virus D(b)NP(366) and D(b)PA(224) epitopes, no significant correlation was found between the duration of CTL contact and the time to phosphatidylserine exposure or loss of membrane integrity. Furthermore, there were minimal indications that transgenic T cells specific for the K(b)OVA(257) epitope (TCR) diversity had any effect. However, when the analysis was repeated with D(b)NP(366) and D(b)PA(224)-specific CTLs recovered directly from the lungs of mice with influenza pneumonia, the lower avidity D(b)NP(366)-specific set was found to elute much more quickly. Shorter contact time may allow individual CTLs to lyse more targets, suggesting that lower TCR/epitope avidity may be more beneficial than higher epitope avidity for cell-mediated immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Microscopy, Video/methods , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Influenza A virus/immunology , Mice , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/physiology
8.
Cell Death Differ ; 15(4): 708-17, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18202705

ABSTRACT

Human GraB (hGraB) preferentially induces apoptosis via Bcl-2-regulated mitochondrial damage but can also directly cleave caspases and caspase substrates in cell-free systems. How hGraB kills cells when it is delivered by cytotoxic lymphocytes (CL) and the contribution of hGraB to CL-induced death is still not clear. We show that primary human natural killer (hNK) cells, which specifically used hGraB to induce target cell death, were able to induce apoptosis of cells whose mitochondria were protected by Bcl-2. Purified hGraB also induced apoptosis of Bcl-2-overexpressing targets but only when delivered at 5- to 10-fold the concentration required to kill cells expressing endogenous Bcl-2. Caspases were critical in this process as inhibition of caspase activity permitted clonogenic survival of Bcl-2-overexpressing cells treated with hGraB or hNK cells but did not protect cells that only expressed endogenous Bcl-2. Our data therefore show that hGraB triggers caspase activation via mitochondria-dependent and mitochondria-independent mechanisms that are activated in a hierarchical manner, and that the combined effects of Bcl-2 and direct caspase inhibition can block cell death induced by hGraB and primary hNK cells.


Subject(s)
Apoptosis , Caspases/metabolism , Granzymes/metabolism , Killer Cells, Natural/enzymology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Secretory Vesicles/enzymology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cell Culture Techniques , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Enzyme Activation , Granzymes/antagonists & inhibitors , Granzymes/genetics , HeLa Cells , Humans , Killer Cells, Natural/drug effects , Mitochondria/enzymology , Mitochondrial Membranes/metabolism , Permeability , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Secretory Vesicles/drug effects , Time Factors , Transfection , X-Linked Inhibitor of Apoptosis Protein/metabolism
10.
Cell Death Differ ; 13(4): 607-18, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16167065

ABSTRACT

Loss of Bid confers clonogenic survival to granzyme B-treated cells, however the exact role of Bid-induced mitochondrial damage--upstream or downstream of caspases--remains controversial. Here we show that direct cleavage of Bid by granzyme B, but not caspases, was required for granzyme B-induced apoptosis. Release of cytochrome c and SMAC, but not AIF or endonuclease G, occurred in the absence of caspase activity and correlated with the onset of apoptosis and loss of clonogenic potential. Loss of mitochondrial trans-membrane potential (DeltaPsim) was also caspase independent, however if caspase activity was blocked the mitochondria regenerated their DeltaPsim. Loss of DeltaPsim was not required for rapid granzyme B-induced apoptosis and regeneration of DeltaPsim following cytochrome c release did not confer clonogenic survival. This functional dissociation of cytochrome c and SMAC release from loss of DeltaPsim demonstrates the essential contribution of Bid upstream of caspase activation during granzyme B-induced apoptosis.


Subject(s)
Apoptosis , Caspases/metabolism , Cytochromes c/metabolism , Mitochondria/physiology , Serine Endopeptidases , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis Inducing Factor/metabolism , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Caspase 3 , Caspase Inhibitors , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Granzymes , HeLa Cells , Humans , Jurkat Cells , Membrane Glycoproteins , Membrane Potentials , Mitochondria/drug effects , Mitochondria/enzymology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Perforin , Pore Forming Cytotoxic Proteins , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , Tumor Stem Cell Assay , Uncoupling Agents/pharmacology
12.
Cell Death Differ ; 10(4): 451-60, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12719722

ABSTRACT

The mechanism of p53-dependent apoptosis is still only partly defined. Using early-passage embryonic fibroblasts (MEF) from wild-type (wt), p53(-/-) and bax(-/-) mice, we observe a p53-dependent translocation of Bax to the mitochondria and a release of mitochondrial Cytochrome c during stress-induced apoptosis. These events proceed independent of zVAD-inhibitable caspase activation, are not prevented by dominant negative FADD (DN-FADD), but are negatively regulated by Mdm-2. Bcl-x(L) expression prevents the release of mitochondrial Cytochrome c and apoptosis, but not Bax translocation. At a single-cell level, enforced expression of p53 is sufficient to induce Bax translocation and Cytochrome c release. Real-time RT-PCR analysis reveals a significant induction of RNA expression of Noxa and Bax in p53(+/+), but not in p53(-/-) MEF. Noxa protein expression becomes detectable prior to Bax translocation, and downregulation of endogenous Noxa by RNA interference protects wt MEF against p53-dependent apoptosis. Hence, in oncogene-expressing MEF p53 induces apoptosis by BH3 protein-dependent caspase activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/genetics , Fibroblasts/enzymology , Mitochondria/enzymology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/deficiency , Tumor Suppressor Protein p53/deficiency , Animals , Carrier Proteins/genetics , Caspases/metabolism , Cells, Cultured , Cytochrome c Group/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Enzyme Inhibitors/pharmacology , Fas-Associated Death Domain Protein , Fetus , Fibroblasts/cytology , Gene Expression Regulation/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Protein Transport/drug effects , Protein Transport/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
13.
Tissue Antigens ; 59(3): 175-83, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12074707

ABSTRACT

The induction of cell death by cytotoxic T-lymphocytes (CTL) or natural killer (NK) cells is one of the main ways by which higher organisms protect themselves from rogue cells, including those infected by a virus, or posing a risk of cancer. Considering the rapidity of viral replication and spread to uninfected cells, CTL and NK are extremely efficient killers. This is at least partly due to the variety of pathways that these cytolytic lymphocytes (CL) can use to ensure the death of a cell. Primarily, CL utilize two independently initiated pathways involving either ligation of death receptors or perforin mediated trafficking of granzyme B to the target cell cytosol to activate a family of death proteases (caspases) in the target cell. The caspases then orchestrate the orderly dismantling of that cell by cleavage of a set of critical substrates. If caspases are inactivated, due either to mutations in proteins that signal their activation or direct inhibition by a viral gene product, CL can utilize a caspase-independent pathway to ensure the death of the target cell. Here we will discuss the mechanisms by which these stellar killers achieve their goal.


Subject(s)
Apoptosis , Caspases/metabolism , Cytotoxicity, Immunologic , T-Lymphocytes, Cytotoxic/immunology , Animals , Apoptosis/physiology , Cell Death/physiology , Granzymes , Killer Cells, Natural/immunology , Membrane Glycoproteins/metabolism , Mice , Models, Immunological , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Tumor Necrosis Factor/metabolism , Serine Endopeptidases/metabolism , Signal Transduction
15.
J Cell Biol ; 153(2): 319-28, 2001 Apr 16.
Article in English | MEDLINE | ID: mdl-11309413

ABSTRACT

During apoptosis, cytochrome c is released into the cytosol as the outer membrane of mitochondria becomes permeable, and this acts to trigger caspase activation. The consequences of this release for mitochondrial metabolism are unclear. Using single-cell analysis, we found that when caspase activity is inhibited, mitochondrial outer membrane permeabilization causes a rapid depolarization of mitochondrial transmembrane potential, which recovers to original levels over the next 30-60 min and is then maintained. After outer membrane permeabilization, mitochondria can use cytoplasmic cytochrome c to maintain mitochondrial transmembrane potential and ATP production. Furthermore, both cytochrome c release and apoptosis proceed normally in cells in which mitochondria have been uncoupled. These studies demonstrate that cytochrome c release does not affect the integrity of the mitochondrial inner membrane and that, in the absence of caspase activation, mitochondrial functions can be maintained after the release of cytochrome c.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cytochrome c Group/metabolism , Intracellular Membranes/metabolism , Mitochondria/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors , Cells, Cultured , Dactinomycin/pharmacology , Fibroblasts/physiology , Flow Cytometry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Membrane Potentials/physiology , Mice , Microscopy, Confocal , Mitochondria/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Uncoupling Agents/pharmacology
16.
J Biol Chem ; 276(15): 12060-7, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11278459

ABSTRACT

Cytotoxic T lymphocytes kill virus-infected and tumor cell targets through the concerted action of proteins contained in cytolytic granules, primarily granzyme B and perforin. Granzyme B, a serine proteinase with substrate specificity similar to the caspase family of apoptotic cysteine proteinases, is capable of cleaving and activating a number of death proteins in target cells. Despite the ability to engage the death pathway at multiple entry points, the preferred mechanism for rapid induction of apoptosis by granzyme B has yet to be clearly established. Here we use time lapse confocal microscopy to demonstrate that mitochondrial cytochrome c release is the primary mode of granzyme B-induced apoptosis and that Bcl-2 is a potent inhibitor of this pivotal event. Caspase activation is not required for cytochrome c release, an activity that correlates with cleavage and activation of Bid, which we have found to be cleaved more readily by granzyme B than either caspase-3 or caspase-8. Bcl-2 blocks the rapid destruction of targets by granzyme B by blocking mitochondrial involvement in the process.


Subject(s)
Apoptosis/drug effects , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , Serine Endopeptidases/pharmacology , Amino Acid Sequence , Cytochrome c Group/metabolism , Enzyme Activation , Granzymes , Humans , Hydrolysis , Jurkat Cells , Kinetics , Molecular Sequence Data
17.
Nat Cell Biol ; 2(3): 156-62, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707086

ABSTRACT

Release of cytochrome c from mitochondria triggers activation of caspase proteases and death of a cell by apoptosis. However, the mechanism and kinetics of cytochrome c release remain unknown. Here we study this event by using green fluorescent protein (GFP)-tagged cytochrome c, and find that the release of cytochrome-c-GFP always precedes exposure of phosphatidylserine and the loss of plasma-membrane integrity - characteristics of apoptotic cells. Once initiated, the release of cytochrome- c-GFP continues until all of the protein is released from all mitochondria in individual cells, within about 5 minutes, regardless of the type or strength of stimulus or the time elapsed since the stimulus was applied. Temperatures ranging from 24 degrees C to 37 degrees C do not change the duration of release, and nor does the addition of caspase inhibitors. Further, we find that the electron-transport chain can maintain the mitochondrial transmembrane potential even after cytochrome c has been released.


Subject(s)
Apoptosis , Cytochrome c Group/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytochrome c Group/genetics , Digitonin/pharmacology , Electron Transport/drug effects , Electron Transport/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Image Processing, Computer-Assisted , Intracellular Membranes/drug effects , Intracellular Membranes/radiation effects , Luminescent Proteins/genetics , Membrane Potentials/drug effects , Mitochondria/metabolism , Oligomycins/pharmacology , Phosphatidylserines/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sodium Azide/pharmacology , Temperature , Ultraviolet Rays
18.
Cell Death Differ ; 7(12): 1192-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11175256

ABSTRACT

Release of cytochrome c from the mitochondria plays an integral role in apoptosis; however, the mechanism by which cytochrome c is released remains one of the conundrums that has occupied the field. Recently, evidence has emerged that the commitment to death may be regulated downstream of cytochrome c release; therefore the mechanism of release must be subtle enough for the cell to recover from this event. In this review, we discuss the evidence that cytochrome c release is mediated by Bcl-2 family proteins in a process that involves only outer membrane permeability but leaves inner membrane energization, protein import function and the ultrastructure of mitochondria intact. Cell Death and Differentiation (2000) 7, 1192 - 1199.


Subject(s)
Apoptosis/physiology , Cytochrome c Group/metabolism , Mitochondria/metabolism , Animals , Cell Membrane Permeability/physiology , Humans , Intracellular Membranes/metabolism , Mitochondria/ultrastructure , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology
19.
Neoplasia ; 2(6): 505-13, 2000.
Article in English | MEDLINE | ID: mdl-11228543

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAID) reduce the risk for cancer, due to their antiproliferative and apoptosis-inducing effects. A critical pathway for apoptosis involves the release of cytochrome c from mitochondria, which then interacts with Apaf-1 to activate caspase proteases that orchestrate cell death. In this study we found that treatment of a human cancer cell line with aspirin induced caspase activation and the apoptotic cell morphology, which was blocked by the caspase inhibitor zVAD-fmk. Further analysis of the mechanism underlying this apoptotic event showed that aspirin induces translocation of Bax to the mitochondria and mitochondrial release of cytochrome into the cytosol. The release of cytochrome c from mitochondria was inhibited by overexpression of the antiapoptotic protein Bcl-2 and cells that lack Apaf-1 were resistant to aspirin-induced apoptosis. These data provide evidence that the release of cytochrome c is an important part of the apoptotic mechanism of aspirin.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Cytochrome c Group/metabolism , Mitochondria/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Apoptotic Protease-Activating Factor 1 , Blotting, Western , Caspase Inhibitors , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Flow Cytometry , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Microscopy, Confocal , Mitochondria/enzymology , Protein Transport , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein
20.
Exp Cell Res ; 251(1): 166-74, 1999 Aug 25.
Article in English | MEDLINE | ID: mdl-10438582

ABSTRACT

Apoptotic cell death involves a series of morphological and biochemical changes orchestrated by activated proteases belonging to the caspase family. Recent studies have suggested that the activation of this process of execution is dependent upon events associated with the loss of mitochondrial inner transmembrane potential (Deltapsi(m)), as a consequence of the formation of the permeability transition (PT) pore. This has led to the proposal that mitochondrial depolarization represents a central irreversible checkpoint in the apoptotic program. Here, we present evidence that HL-60 cells undergo apoptosis in response to the cytotoxic insults of actinomycin-D, etoposide, and staurosporine without showing significant changes in Deltapsi(m). Instead, the loss of Deltapsi(m) could be detected only later in the cell death pathway. In addition, the uncoupling agent CCCP produced an early mitochondrial depolarization in HL-60s but these cells showed few signs of apoptosis up to 8 h after the insult. Furthermore, examination of these cells in response to staurosporine revealed the release of mitochondrial cytochrome c into the cytosol over time, corresponding to caspase activation irrespective of mitochondrial depolarization. In summary, our data suggest that the collapse of Deltapsi(m) as a consequence of PT is not a universal early marker for apoptosis and, moreover, it is not part of the central apoptotic machinery.


Subject(s)
Apoptosis , Intracellular Membranes/physiology , Membrane Potentials/physiology , Mitochondria/physiology , Apoptosis/drug effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Caspase 3 , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Size/drug effects , Cytochrome c Group/metabolism , DNA Fragmentation/drug effects , Dactinomycin/pharmacology , Enzyme Activation/drug effects , Etoposide/pharmacology , Fluorescent Dyes , HL-60 Cells , Humans , Intracellular Membranes/drug effects , Jurkat Cells , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Phosphatidylserines/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Staurosporine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...