Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Biochimie ; 190: 91-110, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34298080

ABSTRACT

Deregulation of apoptosis is associated with various pathologies, such as neurodegenerative disorders at one end of the spectrum and cancer at the other end. Generally speaking, differentiated cells like cardiomyocytes, skeletal myocytes and neurons exhibit low levels of Apaf-1 (Apoptotic protease activating factor 1) protein suggesting that down-regulation of Apaf-1 is an important event contributing to the resistance of these cells to apoptosis. Nonetheless, upregulation of Apaf-1 has not emerged as a common phenomenon in pathologies associated with enhanced neuronal cell death, i.e., neurodegenerative diseases. In cancer, on the other hand, Apaf-1 downregulation is a common phenomenon, which occurs through various mechanisms including mRNA hyper-methylation, gene methylation, Apaf-1 localization in lipid rafts, inhibition by microRNAs, phosphorylation, and interaction with specific inhibitors. Due to the diversity of these mechanisms and involvement of other factors, defining the exact contribution of Apaf-1 to the development of cancer in general and neurodegenerative disorders, in particular, is complicated. The current review is an attempt to provide a comprehensive image of Apaf-1's contribution to the pathologies observed in cancer and neurodegenerative diseases with the emphasis on the therapeutic aspects of Apaf-1 as an important target in these pathologies.


Subject(s)
Apoptotic Protease-Activating Factor 1/metabolism , Neoplasms/etiology , Neurodegenerative Diseases/etiology , Animals , Apoptosomes/drug effects , Apoptotic Protease-Activating Factor 1/agonists , Apoptotic Protease-Activating Factor 1/antagonists & inhibitors , Apoptotic Protease-Activating Factor 1/genetics , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism
2.
Cell Death Dis ; 11(5): 308, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366831

ABSTRACT

Caspase-2, -9, and -3 are reported to control myoblast differentiation into myotubes. This had been previously explained by phosphatidylserine exposure on apoptotic myoblasts inducing differentiation in neighboring cells. Here we show for the first time that caspase-3 is activated in the myoblasts undergoing differentiation. Using RNAi, we also demonstrate that differentiation requires both cytochrome c and Apaf-1, and by using a new pharmacological approach, we show that apoptosome formation is required. We also show that Bid, whose cleavage links caspase-2 to the mitochondrial death pathway, was required for differentiation, and that the caspase cleavage product, tBid, was generated during differentiation. Taken together, these data suggest that myoblast differentiation requires caspase-2 activation of the mitochondrial death pathway, and that this occurs in the cells that differentiate. Our data also reveal a hierarchy of caspases in differentiation with caspase-2 upstream of apoptosome activation, and exerting a more profound control of differentiation, while caspases downstream of the apoptosome primarily control cell fusion.


Subject(s)
Apoptosomes/metabolism , Caspase 3/metabolism , Cell Differentiation , Muscle Fibers, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/enzymology , Animals , Apoptosis/drug effects , Apoptosomes/drug effects , Apoptotic Protease-Activating Factor 1/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 2/metabolism , Caspase Inhibitors/pharmacology , Cell Differentiation/drug effects , Cell Fusion , Cell Line , Cyclohexanones/pharmacology , Cytochromes c/metabolism , Enzyme Activation/drug effects , Gene Knockdown Techniques , Humans , Mice , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , RNA, Small Interfering/metabolism
3.
FEBS Open Bio ; 9(7): 1194-1203, 2019 07.
Article in English | MEDLINE | ID: mdl-31033240

ABSTRACT

The expense and time required for in vivo reproductive and developmental toxicity studies have driven the development of in vitro alternatives. Here, we used a new in vitro split luciferase-based assay to screen a library of 177 toxicants for inhibitors of apoptosome formation. The apoptosome contains seven Apoptotic Protease-Activating Factor-1 (Apaf-1) molecules and induces cell death by activating caspase-9. Apaf-1-dependent caspase activation also plays an important role in CNS development and spermatogenesis. In the in vitro assay, Apaf-1 fused to an N-terminal fragment of luciferase binds to Apaf-1 fused to a C-terminal fragment of luciferase and reconstitutes luciferase activity. Our assay indicated that pentachlorophenol (PCP) inhibits apoptosome formation, and further investigation revealed that PCP binds to cytochrome c. PCP is a wood preservative that reduces male fertility by ill-defined mechanisms. Although the data show that PCP inhibited apoptosome formation, the concentration required suggests that other mechanisms may be more important for PCP's effects on spermatogenesis. Nonetheless, the data demonstrate the utility of the new assay in identifying apoptosome inhibitors, and we suggest that the assay may be useful in screening for reproductive and developmental toxicants.


Subject(s)
Apoptosomes/drug effects , Pentachlorophenol/toxicity , Toxicity Tests/methods , Apoptosis/drug effects , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Cell Death , Cytochromes c/metabolism , HEK293 Cells , Humans , Luciferases/metabolism , Pentachlorophenol/pharmacology , Signal Transduction , Small Molecule Libraries
4.
J Cell Biochem ; 120(6): 10662-10669, 2019 06.
Article in English | MEDLINE | ID: mdl-30652348

ABSTRACT

Earlier studies showed that the oxidant menadione (MD) induces apoptosis in certain cells and also has anticancer effects. Most of these studies emphasized the role of the mitochondria in this process. However, the engagement of other organelles is less known. Particularly, the role of lysosomes and their proteolytic system, which participates in apoptotic cell death, is still unclear. The aim of this study was to investigate the role of lysosomal cathepsins on molecular signaling in MD-induced apoptosis in U937 cells. MD treatment induced translocation of cysteine cathepsins B, C, and S, and aspartic cathepsin D. Once in the cytosol, some cathepsins cleaved the proapoptotic molecule, Bid, in a process that was completely prevented by E64d, a general inhibitor of cysteine cathepsins, and partially prevented by the pancaspase inhibitor, z-VAD-fmk. Upon loss of the mitochondrial membrane potential, apoptosome activation led to caspase-9 processing, activation of caspase-3-like caspases, and poly (ADP-ribose) polymerase cleavage. Notably, the endogenous protein inhibitor, stefin B, was degraded by cathepsin D and caspases. This process was prevented by z-VAD-fmk, and partially by pepstatin A-penetratin. These findings suggest that the cleaved Bid protein acts as an amplifier of apoptotic signaling through mitochondria, thus enhancing the activity of cysteine cathepsins following stefin B degradation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/genetics , Cystatin B/genetics , Gene Expression Regulation, Neoplastic , Lysosomes/drug effects , Vitamin K 3/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/genetics , Apoptosomes/drug effects , Apoptosomes/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin C/antagonists & inhibitors , Cathepsin C/genetics , Cathepsin C/metabolism , Cathepsin D/antagonists & inhibitors , Cathepsin D/genetics , Cathepsin D/metabolism , Cathepsins/antagonists & inhibitors , Cathepsins/genetics , Cathepsins/metabolism , Cystatin B/metabolism , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Lysosomes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Pepstatins/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Protease Inhibitors/pharmacology , Proteolysis/drug effects , Signal Transduction , U937 Cells
5.
Pak J Pharm Sci ; 31(6(Special)): 2787-2790, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30630785

ABSTRACT

Hyaluronic acid (HA) is used to aid tissue repair and is a characterized inhibitor of TRPV1 channels. In this study, we investigated the effects of HA on lidocaine induced neurotoxicity and its mechanism of action. U87-MG cells with low (U87-MG-shTRPV1) or high (U87-MG-TRPV1) TRPV1 expression were studied. The control group was treated with lidocaine. The experimental group was treated with lidocaine and HA. Flow cytometry was used to assess the intracellular calcium concentration ([Ca2+] i) and cell apoptosis. Cell viability was detected by MTT assays. Compared to the control group, [Ca2+]i of U87-MG-TRPV1 and U87-MG cells were lower at T3, T4 and T5 (p < 0.05), apoptosis rates of U87-MG and U87-MG-TRPV1 cells were lower (p<0.05), and the cell viability of U87-MG and U87-MG-TRPV1 cells were higher in the experimental group (p<0.05). HA reduces the toxic damage of lidocaine through blocking Ca2+ influx through TRPV1 channels, preventing Ca2+ overload, leading to nerve cell protection.


Subject(s)
Hyaluronic Acid/pharmacology , Neurotoxicity Syndromes/prevention & control , Apoptosomes/drug effects , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Humans , Lidocaine , TRPV Cation Channels/biosynthesis
6.
Biochem Biophys Res Commun ; 465(2): 299-304, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26265044

ABSTRACT

Heart diseases such as myocardial infarction (MI) can damage individual cardiomyocytes, leading to the activation of cell death programs. The most scrutinized type of cell death in the heart is apoptosis, and one of the key events during the propagation of apoptotic signaling is the formation of apoptosomes, which relay apoptotic signals by activating caspase-9. As one of the major components of apoptosomes, apoptotic protease activating factor 1 (Apaf-1) facilitates the formation of apoptosomes containing cytochrome c (Cyto-c) and deoxyadenosine triphosphate (dATP). Thus, it may be possible to suppress the activation of the apoptotic program by down-regulating the expression of Apaf-1 using miRNAs. To validate this hypothesis, we selected a number of candidate miRNAs that were expected to target Apaf-1 based on miRNA target prediction databases. Among these candidate miRNAs, we empirically identified miR-17 as a novel Apaf-1-targeting miRNA. The delivery of exogenous miR-17 suppressed Apaf-1 expression and consequently attenuated formation of the apoptosome complex containing caspase-9, as demonstrated by co-immunoprecipitation and immunocytochemistry. Furthermore, miR-17 suppressed the cleavage of procaspase-9 and the subsequent activation of caspase-3, which is downstream of activated caspase-9. Cell viability tests also indicated that miR-17 pretreatment significantly prevented the norepinephrine-induced apoptosis of cardiomyocytes, suggesting that down-regulation of apoptosome formation may be an effective strategy to prevent cellular apoptosis. These results demonstrate the potential of miR-17 as an effective anti-apoptotic agent.


Subject(s)
Apoptosis/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosomes/drug effects , Apoptosomes/genetics , Apoptotic Protease-Activating Factor 1/genetics , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cytochromes c/genetics , Cytochromes c/metabolism , Deoxyadenine Nucleotides/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Norepinephrine/pharmacology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Signal Transduction , Transfection
7.
J Nutr Biochem ; 23(3): 245-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21497497

ABSTRACT

Immobilization is characterized by activation of the ubiquitin (Ub)-proteasome-dependent proteolytic system (UPS) and of the mitochondrial apoptotic pathway. Increased oxidative stress and inflammatory response occur in immobilized skeletal muscles. Curcumin exhibits antioxidant and anti-inflammatory properties, blocked proteasome activation in intact animals, and may favor skeletal muscle regeneration. We therefore measured the effects of curcumin on immobilization-induced muscle atrophy and subsequent recovery. Rats were subjected to hindlimb immobilization for 8 days (I8) and allowed to recover for 10 days (R10). Fifty percent of the rats were injected daily with either curcumin or vehicle. Proteolytic and apoptotic pathways were studied in gastrocnemius muscles. Curcumin treatment prevented the enhanced proteasome chymotrypsin-like activity and the trend toward increased caspase-9-associated apoptosome activity at I8 in immobilized muscles. By contrast, the increase of these two activities was blunted by curcumin at R10. Curcumin did not reduce muscle atrophy at I8 but improved muscle recovery at R10 and the cross-sectional area of muscle fibers of immobilized muscles. Curcumin reduced the increased protein levels of Smac/DIABLO induced by immobilization and enhanced the elevation of X-linked inhibitory apoptotic protein levels at R10. Ub-conjugate levels and caspase-3 activity increased at I8 and were normalized at R10 without being affected by curcumin treatment. Altogether, the data show that curcumin treatment improved recovery during reloading. The effect of curcumin during the atrophic phase on proteasome activities may facilitate the initiation of muscle recovery after reloading. These data also suggest that this compound may favor the initial steps of muscle regeneration.


Subject(s)
Apoptosomes/drug effects , Curcumin/pharmacology , Muscle, Skeletal/drug effects , Plant Extracts/pharmacology , Proteasome Endopeptidase Complex/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Apoptosomes/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Hindlimb Suspension , Male , Mitochondria/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/drug therapy , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Wistar , Regeneration/drug effects , Up-Regulation
8.
Toxicol Sci ; 122(1): 64-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21527774

ABSTRACT

Electrophile-mediated post-translational modifications (PTMs) are known to cause tissue toxicities and disease progression. These effects are mediated via site-specific modifications and structural disruptions associated with such modifications. 1,4-Benzoquinone (BQ) and its quinone-thioether metabolites are electrophiles that elicit their toxicity via protein arylation and the generation of reactive oxygen species. Site-specific BQ-lysine adducts are found on residues in cytochrome c that are necessary for protein-protein interactions, and these adducts contribute to interferences in its ability to facilitate apoptosome formation. To further characterize the structural and functional impact of these BQ-mediated PTMs, the original mixture of BQ-adducted cytochrome c was fractionated by liquid isoelectric focusing to provide various fractions of BQ-adducted cytochrome c species devoid of the native protein. The fractionation process separates samples based on their isoelectric point (pI), and because BQ adducts form predominantly on lysine residues, increased numbers of BQ adducts on cytochrome c correlate with a lower protein pI. Each fraction was analyzed for structural changes, and each was also assayed for the ability to support apoptosome-mediated activation of caspase-3. Circular dichroism revealed that several of the BQ-adducted cytochrome c species maintained a slightly more rigid structure in comparison to native cytochrome c. BQ-adducted cytochrome c also failed to activate caspase-3, with increasing numbers of BQ-lysine adducts corresponding to a greater inability to activate the apoptosome. In summary, the specific site of the BQ-lysine adducts, and the nature of the adduct, are important determinants of the subsequent structural changes to cytochrome c. In particular, adducts at sites necessary for protein-protein interactions interfere with the proapoptotic function of cytochrome c.


Subject(s)
Apoptosomes/drug effects , Apoptosomes/metabolism , Benzoquinones/toxicity , Cytochromes c/chemistry , DNA Adducts , Lysine/metabolism , Animals , Benzoquinones/chemistry , Caspase 3/metabolism , Chromatography, Liquid , Circular Dichroism/methods , Horses , Isoelectric Focusing/methods , Models, Molecular , Protein Conformation , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary , Tandem Mass Spectrometry
9.
PLoS One ; 6(12): e29058, 2011.
Article in English | MEDLINE | ID: mdl-22216167

ABSTRACT

Alternative splicing of caspase-3 produces a short isoform caspase-3s that antagonizes caspase-3 apoptotic activity. However, the mechanism of apoptosis inhibition by caspase-3s remains unknown. Here we show that exogenous caspase-3 sensitizes MCF-7 and HBL100 breast cancers cells to chemotherapeutic treatments such as etoposide and methotrexate whereas co-transfection with caspase-3s strongly inhibits etoposide and methotrexate-induced apoptosis underlying thus the anti-apoptotic role of caspase-3s. In caspase-3 transfected cells, lamin-A and α-fodrin were cleaved when caspase-3 was activated by etoposide or methotrexate. When caspase-3s was co-transfected, this cleavage was strongly reduced. Depletion of caspase-3 by RNA interference in HBL100 containing endogenous caspase-3s caused reduction in etoposide and methotrexate-induced apoptosis, whereas the depletion of caspase-3s sensitized cells to chemotherapy. In the presence of caspase-3s, a lack of interaction between caspase-3 and caspase-9 was observed. Immunoprecipitation assays showed that caspase-3s binds the pro-forms of caspase-3. This result suggested that the absence of interaction with caspase-9 when both variants of caspase-3 are present contribute to block the apoptosome assembly and inhibit apoptosis. These data support that caspases-3s negatively interferes with caspase-3 activation and apoptosis in breast cancer, and that it can play key roles in the modulation of response to chemotherapeutic treatments.


Subject(s)
Apoptosis/drug effects , Apoptosomes/drug effects , Caspase 3/metabolism , Isoenzymes/metabolism , Amino Acid Sequence , Base Sequence , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Caspase 3/chemistry , Cell Line, Tumor , DNA Primers , Enzyme Activation , Humans , Isoenzymes/chemistry , Models, Molecular , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid
10.
Biochemistry ; 49(9): 1923-30, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20121050

ABSTRACT

Structurally diverse small molecules, including 5-(2-benzofuryl)-4-phenyl-1,2,4-triazole-3-thiol (BETT), have been identified via high-throughput screening as activators of caspase-3 in HeLa cell extracts. However, little is known about their mechanism of action. In this study, we investigate how BETT regulates prothymosin alpha (ProT), a nuclear protein previously shown to play essential roles in apoptosis. We first showed that Apaf-1 is the direct target protein of BETT. We further demonstrated that BETT relieved ProT-mediated inhibition of apoptosome formation by blocking the interaction between Apaf-1 and ProT. Using two-dimensional (1)H-(15)N heteronuclear single-quantum correlation (HSQC) experiments, we were also able to examine the interaction between Apaf-1 and (15)N-labeled ProT alpha. Furthermore, we were able to reconstitute the entire caspase-3 activation pathway using purified ProT, Apaf-1, procaspase-9, procaspase-3, Hsp70, cytochrome c, PHAPI, CAS, and regulatory compounds to mimic stress-induced apoptosis in vitro. Together, these studies would lead to novel and specific methods for the prevention, diagnosis, and treatment of human cancer.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/antagonists & inhibitors , Apoptotic Protease-Activating Factor 1/metabolism , Protein Precursors/antagonists & inhibitors , Protein Precursors/physiology , Thymosin/analogs & derivatives , Triazoles/pharmacology , Animals , Apoptosis/drug effects , Apoptosomes/chemistry , Apoptosomes/drug effects , Apoptotic Protease-Activating Factor 1/chemistry , Caspase 3/metabolism , Caspase Inhibitors , Cell Line, Tumor , Drug Delivery Systems , HeLa Cells , Humans , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondria/physiology , Protein Binding , Protein Precursors/genetics , Protein Precursors/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera , Thymosin/antagonists & inhibitors , Thymosin/genetics , Thymosin/metabolism , Thymosin/physiology
11.
Apoptosis ; 15(4): 401-11, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20091232

ABSTRACT

In the past few years, overwhelming evidence has accrued that a high level of expression of the protein neuroglobin protects neurons in vitro, in animal models, and in humans, against cell death associated with hypoxic and amyloid insult. However, until now, the exact mechanism of neuroglobin's protective action has not been determined. Using cell biology and biochemical approaches we demonstrate that neuroglobin inhibits the intrinsic pathway of apoptosis in vitro and intervenes in activation of pro-caspase 9 by interaction with cytochrome c. Using systems level information of the apoptotic signalling reactions we have developed a quantitative model of neuroglobin inhibition of apoptosis, which simulates neuroglobin blocking of apoptosome formation at a single cell level. Furthermore, this model allows us to explore the effect of neuroglobin in conditions not easily accessible to experimental study. We found that the protection of neurons by neuroglobin is very concentration sensitive. The impact of neuroglobin may arise from both its binding to cytochrome c and its subsequent redox reaction, although the binding alone is sufficient to block pro-caspase 9 activation. These data provides an explanation the action of neuroglobin in the protection of nerve cells from unwanted apoptosis.


Subject(s)
Apoptosis , Cytoprotection , Globins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/metabolism , Signal Transduction , Apoptosis/drug effects , Apoptosomes/drug effects , Apoptosomes/metabolism , Benzopyrans/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Computational Biology , Cytochromes c/metabolism , Cytoprotection/drug effects , Cytosol/drug effects , Cytosol/metabolism , Enzyme Activation/drug effects , Humans , Models, Biological , Models, Molecular , Neuroglobin , Neurons/drug effects , Neurons/enzymology , Nitriles/pharmacology , Oxidation-Reduction/drug effects , Protein Structure, Secondary , Signal Transduction/drug effects
12.
Free Radic Biol Med ; 45(12): 1723-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18926904

ABSTRACT

A proposed mechanism for the cardiotoxicity of doxorubicin (DOX) involves apoptosis in cardiomyocytes. In the study described here, we investigated the molecular basis for the differences in DOX-induced toxicity in adult rat cardiomyocytes (ARCM), neonatal rat cardiomyocytes (NRCM), and rat embryonic H9c2 cardiomyoblasts. Activation of caspase-9 and -3 was considerably lower in DOX-treated ARCM as compared with NRCM and H9c2 cardiomyoblasts. Addition of cytochrome c caused the activation of caspase-9 and -3 in permeabilized NRCM and H9c2 cardiomyoblasts but not in permeabilized ARCM. Expression of proapoptotic proteins, apoptotic protease activating factor-1 (Apaf1), and procaspase-9 was significantly lower, and abundance of antiapoptotic X-linked inhibitor of apoptosis protein (XIAP) was higher in ARCM, as compared with immature cardiac cells. Despite the abundance of XIAP in ARCM, its role in the inhibition of apoptosome function was dismissed, as second mitochondria-derived activator of caspases (Smac)-N7 peptide, had no effect on caspase activation in response to cytochrome c in these cells. Adenoviral expression of Apaf1 exacerbated the activation of caspase-9 and -3 in DOX-treated NRCM, but did not increase their activities in DOX-treated ARCM. This finding points to a major difference in the apoptotic signaling between immature and adult cardiomyocytes. The mitochondrial apoptotic pathway is limited in ARCM treated with DOX.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosomes/drug effects , Doxorubicin/pharmacology , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects , Adenoviridae/genetics , Animals , Animals, Newborn , Apoptosis Regulatory Proteins , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Carrier Proteins/metabolism , Caspases/metabolism , Cytochromes c/metabolism , Enzyme Activation/drug effects , Immunoblotting , Male , Mitochondrial Proteins/metabolism , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
13.
Cell Death Differ ; 15(11): 1712-22, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18617896

ABSTRACT

Bcr-Abl tyrosine kinase (TK) inhibitors are promising therapeutic agents for Bcr-Abl-positive (Bcr-Abl(+)) leukemias. Although they are known to promote caspase-mediated apoptosis, it remains unclear whether caspase-independent cell death-inducing mechanisms are also triggered. Here we demonstrated that INNO-406, a second-generation Bcr-Abl TK inhibitor, induces programmed cell death (PCD) in chronic myelogenous leukemia (CML) cell lines through both caspase-mediated and caspase-independent pathways. The latter pathways include caspase-independent apoptosis (CIA) and necrosis-like cell death (CIND), and the cell lines varied regarding which mechanism was elicited upon INNO-406 treatment. We also observed that the propensity toward CIA or CIND in cells was strongly associated with cellular dependency on apoptosome-mediated caspase activity. Cells that undergo CIND have a high apoptosome activity potential whereas cells that undergo CIA tend to have a lower potential. Moreover, we found that INNO-406 promotes autophagy. When autophagy was inhibited with chloroquine or gene knockdown of beclin1 by shRNA, INNO-406-induced cell death was enhanced, which indicates that the autophagic response of the tumor cells is protective. These findings suggest new insights into the biology and therapy of Bcr-Abl(+) leukemias.


Subject(s)
Autophagy/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosomes/drug effects , Apoptosomes/metabolism , Caspases/metabolism , Cell Line, Tumor , Chloroquine/pharmacology , Cytoprotection/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Male , Mice , Mice, SCID , Xenograft Model Antitumor Assays
14.
Exp Cell Res ; 314(3): 651-67, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18166172

ABSTRACT

Constitutively expressed HABP1 in normal murine fibroblast cell line induces growth perturbation, morphological abnormalities along with initiation of apoptosis. Here, we demonstrate that though HABP1 accumulation started in mitochondria from 48 hr of growth, induction of apoptosis with the release of cytochrome c and apoptosome complex formation occurred only after 60 hr. This mitochondrial dysfunction was due to gradual increase in ROS generation in HABP1 overexpressing cells. Along with ROS generation, increased Ca 2+ influx in mitochondria leading to drop in membrane potential was evident. Interestingly, upon expression of HABP1, the respiratory chain complex I was shown to be significantly inhibited. Electronmicrograph confirmed defective mitochondrial ultrastructure. The reduction in oxidant generation and drop in apoptotic cell population accomplished by disruption of HABP1 expression, corroborating the fact that excess ROS generation in HABP1 overexpressing cells leading to apoptosis was due to mitochondrial HABP1 accumulation.


Subject(s)
Apoptosis/physiology , Fibroblasts/metabolism , Hyaluronan Receptors/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Apoptosomes/drug effects , Apoptosomes/metabolism , Cell Line , Cytochromes c/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Proteins , Oxidative Stress/drug effects , Reactive Oxygen Species/pharmacology , Time Factors
15.
Pharm Res ; 25(4): 740-51, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17674158

ABSTRACT

Apoptosis plays a crucial role in tissue homeostasis, development and many diseases. The relevance of Apaf1, the molecular core of apoptosome, has been underlined in mitochondria-dependent apoptosis, which according to a growing body of evidence, is involved in various pathologies where the equilibrium of life-and-death is dysregulated, such as heart attack, stroke, liver failure, cancer and autoimmune diseases. Consequently, great interest has emerged in devising therapeutic strategies for regulating the key molecules involved in the life-and-death decision. Here we review recent progress in apoptosis-based pharmacological therapies and, in particular, we point out a possible role of the apoptosome as an emerging and promising pharmacological target.


Subject(s)
Apoptosis/drug effects , Apoptosomes/drug effects , Caspases/metabolism , Drug Design , Signal Transduction/drug effects , Animals , Drug Evaluation, Preclinical , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism
16.
Biochemistry ; 46(39): 11090-100, 2007 Oct 02.
Article in English | MEDLINE | ID: mdl-17824617

ABSTRACT

Electrophiles generated endogenously, or via the metabolic bioactivation of drugs and other environmental chemicals, are capable of binding to a variety of nucleophilic sites within proteins. Factors that determine site selective susceptibility to electrophile-mediated post-translational modifications, and the consequences of such alterations, remain largely unknown. To identify and characterize chemical-mediated protein adducts, electrophiles with known toxicity were utilized. Hydroquinone, and its mercapturic acid pathway metabolites, cause renal proximal tubular cell necrosis and nephrocarcinogenicity in rats. The adverse effects of HQ and its thioether metabolites are in part a consequence of their oxidation to the corresponding electrophilic 1,4-benzoquinones (BQ). We now report that BQ and 2-(N-acetylcystein-S-yl)benzoquinone (NAC-BQ) preferentially bind to solvent-exposed lysine-rich regions within cytochrome c. Furthermore, we have identified specific glutamic acid residues within cytochrome c as novel sites of NAC-BQ adduction. The microenvironment at the site of adduction governs both the initial specificity and the structure of the final adduct. The solvent accessibility and local pKa of the adducted and neighboring amino acids contribute to the selectivity of adduction. Postadduction chemistry subsequently alters the nature of the final adduct. Using molecular modeling, the impact of BQ and NAC-BQ adduction on cytochrome c was visualized, revealing the spatial rearrangement of critical residues necessary for protein-protein interactions. Consequently, BQ-adducted cytochrome c fails to initiate caspase-3 activation in native lysates and also inhibits Apaf-1 oligomerization into an apoptosome complex in a purely reconstituted system. In summary, a combination of mass spectroscopic, molecular modeling, and biochemical approaches confirms that electrophile-protein adducts produce structural alterations that influence biological function.


Subject(s)
Apoptosomes/metabolism , Benzoquinones/chemistry , Cytochromes c/chemistry , Acetylcysteine/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosomes/drug effects , Benzoquinones/pharmacology , Caspase 3/chemistry , Caspase 3/metabolism , Caspase 9/chemistry , Caspase 9/metabolism , Cell Line, Tumor , Chromatography, Liquid , Circular Dichroism , Cytochromes c/metabolism , Horses , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Binding/drug effects , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
17.
Anticancer Res ; 27(4B): 2175-83, 2007.
Article in English | MEDLINE | ID: mdl-17695501

ABSTRACT

The novel cyclopenta[b]benzofuran, silvestrol, isolated from the fruits and twigs of Aglaia foveolata, has been found to exhibit very potent in vitro cytotoxic activity against several human cancer cell lines. Furthermore, it was active in the in vivo P388 murine leukemia model. In this study, the mechanism of cytotoxicity mediated by silvestrol in the LNCaP (hormone-dependent human prostate cancer) cell line was investigated. Silvestrol induced an apoptotic response, disrupted the mitochondrial trans-membrane potential and caused cytochrome c release into the cytoplasm. Immunoblot analysis indicated that, at the protein level, silvestrol produced an increase of Bcl-xl phosphorylation with a concomitant increase of bak. Furthermore, caspase-2, -9 and -10 appeared to be involved in silvestrol-mediated apoptosis. In contrast, the involvement of caspase-3 and -7 was not detected, either by immunoblot or caspase-3/-7-like activity analysis, indicating that these pathways do not play a crucial role in silvestrol-induced apoptosis. To investigate the relative contribution of the caspases, inhibition of apoptosis with four different cell-permeable inhibitors was studied (Boc-D-Fmk, Z-VDVAD-FMK Z-LEHD-FMK and Z-AEVD-FMK). Only the general caspase inhibitor, Boc-D-Fmk, completely inhibited the formation of apoptotic bodies. In contrast, caspase-2 and caspase-9 selective inhibitors induced about a 40% reduced apoptotic response, whereas the caspase-10 selective inhibitor caused about a 60% reduction in apoptosis compared to silvestrol only treated cells. Taken together, the studies described herein demonstrate the involvement of the apoptosome/mitochondrial pathway and suggest the possibility that silvestrol may also trigger the extrinsic pathway of programmed cell death signaling in tumor cells.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Triterpenes/pharmacology , Aglaia/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Apoptosomes/drug effects , Apoptosomes/physiology , Benzyl Compounds/pharmacology , Cell Line, Tumor , Drug Interactions , Enzyme Activation , Humans , Hydrocarbons, Fluorinated/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/drug effects , Mitochondria/physiology , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology
18.
Cancer Biol Ther ; 6(2): 209-17, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17224646

ABSTRACT

The apoptosome is a multiprotein complex mediating the mitochondrial pathway of cell death. Its importance during development has been clearly demonstrated by knocking out key genes in mouse. APAF1 is the core protein of the apoptosome and its dosage is also critical in various cancer types, i.e., melanoma, germ line tumor, gastrointestinal cancer and B-type chronic lymphocytic leukemia. This is generally due to inactivation of the APAF1 locus by epigenetic phenomena or by activity of promoter regulators. We investigated the putative roles of the apoptosome in pancreatic ductal adenocarcinoma (PDAC). We found that both APAF1 mRNA and protein are dysregulated in human PDAC samples. Similarly, several PDAC cell lines exhibited variable levels of both APAF1 protein and mRNA. The response to cell death induction and its biochemical features were assessed by treatment of each line with commonly used chemotherapeutic agents. We found that the apoptosome pathway was not functional in most cell lines upon cytochrome c release from mitochondria. In addition, we restored APAF1 and Caspase-9 dosage in Panc-1 cells, where the apoptosome is downregulated, by overexpressing the murine cDNA of the two molecules, and we improved the death response to chemotherapeutic agents.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Caspase 9/metabolism , Pancreatic Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosomes/drug effects , Apoptotic Protease-Activating Factor 1/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Caspase 9/drug effects , Cell Death , Cell Line, Tumor , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Pancreatic Neoplasms/drug therapy
19.
Mol Cell ; 25(2): 181-92, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17244527

ABSTRACT

Apaf-1 plays an essential role in apoptosis. In the presence of cytochrome c and dATP, Apaf-1 assembles into an oligomeric apoptosome, which is responsible for the activation of procaspase-9 and the maintenance of the enzymatic activity of the processed caspase-9. Regulation of apoptosome assembly by other cellular factors is poorly understood. Here we report that physiological concentrations of calcium ion negatively affect the assembly of apoptosome by inhibiting nucleotide exchange in the monomeric, autoinhibited Apaf-1 protein. Consequently, calcium blocks the ability of Apaf-1 to activate caspase-9. These observations suggest an important role of calcium homeostasis on the Apaf-1-dependent apoptotic pathway.


Subject(s)
Apoptosomes/drug effects , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Calcium/pharmacology , Adenine Nucleotides/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/genetics , Calcium/metabolism , Caspase 9/metabolism , Enzyme Activation/drug effects , Homeostasis , Humans , In Vitro Techniques , Models, Biological , Protein Conformation/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Oncogene ; 25(58): 7635-45, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-16767158

ABSTRACT

Anticancer drugs can induce tumor cell death by caspase-dependent apoptosis. The observation that procaspase-10 expression decreased in leukemic cells from acute myeloblastic leukemia patients at first relapse led us to explore the role of caspase-10 in cytotoxic drug-induced apoptosis. We show that caspase-10 is activated in etoposide-treated cells in a dose- and time-dependent manner. A caspase-10 peptide inhibitor, a caspase-10 dominant-negative mutant or a small interfering RNA (siRNA)-mediated downregulation of the enzyme negatively interfere with drug-induced cell death and caspase-2, -3, -8 and -9 activation. The extrinsic pathway to apoptosis is not involved in drug-induced caspase-10 activation that occurs downstream of Bax redistribution to mitochondria and cytochrome c release from this organelle. siRNA-mediated downregulation of Apaf-1 prevents etoposide-mediated activation of caspase-10. In a cell-free assay, cytochrome c and dATP treatment of cell extracts after immunodepletion of either caspase-3 or caspase-9 indicates that caspase-10 is activated downstream of caspase-9. Then, caspase-10 is involved in a feedback amplification loop that amplifies caspase-9 and -3 activities. Altogether, these data indicate an active role for caspase-10 in cytotoxic drug-induced tumor cell death, downstream of the mitochondria.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Caspase 10/physiology , Etoposide/pharmacology , Signal Transduction , Apoptosomes/drug effects , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Leukemia, Myeloid, Acute/diagnosis , Mitochondria/metabolism , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL