Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biochimie ; 180: 23-29, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33132160

ABSTRACT

Split luciferase complementary assay has been used to investigate the effect of WD domain deletion on Apaf-1 oligomerization. Apaf-1 is an adaptor molecule in formation of apoptosome that activates caspase-9, an activation that is a key event in the mitochondrial cell death pathway. Structural studies suggest that normally Apaf-1 is held in an inactive conformation by intramolecular interactions between Apaf-1's nucleotide binding domain and one of its WD40 domains (WD1). In the prevailing model of Apaf-1 activation, cytochrome c binds to sites in WD1 and in Apaf-1's second WD40 domain (WD2), moving WD1 and WD2 closer together and rotating WD1 away from the nucleotide binding domain. This allows Apaf-1 to bind dATP or ATP and to form the apoptosome, which activates caspase-9. This model predicts that cytochrome c binding to both WD domains is necessary for apoptosome formation and that an Apaf-1 with only WD1 will be locked in an inactive conformation that cannot be activated by cytochrome c. Here we investigated the effect of removing one WD domain (Apaf-1 1-921) on Apaf-1 interactions and caspase activation. Apaf-1 1-921 could not activate caspase-9, even in the presence of cytochrome c. These data show that a single WD domain is sufficient to lock Apaf-1 in an inactive state and this state cannot be altered by cytochrome c.


Subject(s)
Apoptosomes/chemistry , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , WD40 Repeats/physiology , Apoptotic Protease-Activating Factor 1/genetics , Caspase 3/metabolism , Caspase 9/metabolism , Cytochromes c/metabolism , Deoxyadenine Nucleotides/metabolism , Enzyme Activation , HEK293 Cells , Humans , Luciferases/metabolism , Luminescent Measurements/methods , Mutation/genetics , Protein Binding , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
2.
Biochim Biophys Acta Mol Cell Res ; 1867(1): 118573, 2020 01.
Article in English | MEDLINE | ID: mdl-31678591

ABSTRACT

Cytochrome c (Cyt c) released from mitochondria interacts with Apaf-1 to form the heptameric apoptosome, which initiates the caspase cascade to execute apoptosis. Although lysine residue at 72 (K72) of Cyt c plays an important role in the Cyt c-Apaf-1 interaction, the underlying mechanism of interaction between Cyt c and Apaf-1 is still not clearly defined. Here we identified multiple lysine residues including K72, which are also known to interact with ATP, to play a key role in Cyt c-Apaf-1 interaction. Mutation of these lysine residues abrogates the apoptosome formation causing inhibition of caspase activation. Using in-silico molecular docking, we have identified Cyt c-binding interface on Apaf-1. Although mutant Cyt c shows higher affinity for Apaf-1, the presence of Cyt c-WT restores the apoptosome activity. ATP addition modulates only mutant Cyt c binding to Apaf-1 but not WT Cyt c binding to Apaf-1. Using TCGA and cBioPortal, we identified multiple mutations in both Apaf-1 and Cyt c that are predicted to interfere with apoptosome assembly. We also demonstrate that transcript levels of various enzymes involved with dATP or ATP synthesis are increased in various cancers. Silencing of nucleotide metabolizing enzymes such as ribonucleotide reductase subunit M1 (RRM1) and ATP-producing glycolytic enzymes PKM2 attenuated ATP production and enhanced caspase activation. These findings suggest important role for lysine residues of Cyt c and nucleotides in the regulation of apoptosome-dependent apoptotic cell death as well as demonstrate how these mutations and nucleotides may have a pivotal role in human diseases such as cancer.


Subject(s)
Apoptosomes/physiology , Cytochromes c/chemistry , Molecular Docking Simulation , Neoplasms/pathology , Nucleotides/chemistry , Alanine/chemistry , Alanine/genetics , Amino Acid Substitution , Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cytochromes c/genetics , Cytochromes c/metabolism , Female , Humans , Lysine/chemistry , Lysine/genetics , Male , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/physiology , Neoplasms/genetics , Neoplasms/metabolism , Nucleotides/metabolism , PC-3 Cells , Protein Binding/genetics , Protein Interaction Mapping , Protein Multimerization/genetics , Signal Transduction/genetics
3.
Arch Biochem Biophys ; 642: 46-51, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29410086

ABSTRACT

Apaf-1 is a cytosolic multi-domain protein in the apoptosis regulatory network. When cytochrome c releases from mitochondria; it binds to WD-40 repeats of Apaf-1 molecule and induces oligomerization of Apaf-1. Here in, a split luciferase assay was used to compare apoptosome formation in cell-free and cell-based systems. This assay uses Apaf-1 tagged with either N-terminal fragment or C-terminal fragment of P. pyralis luciferase. In cell based-system, the apoptosome formation is induced inside the cells which express Apaf-1 tagged with complementary fragments of luciferase while in cell-free system, the apoptosome formation is induced in extracts of the cells. In cell-free system, cytochrome c dependent luciferase activity was observed with full length Apaf-1. However, luciferase activity due to apoptosome formation was much higher in cell based system compared to cell-free system. The truncated Apaf-1 which lacks WD-40 repeats (ΔApaf-1) interacted with endogenous Apaf-1 in a different fashion compared to native form as confirmed by different retention time of eluate in gel filtration and binding to affinity column. The interactions between endogenous Apaf-1 and ΔApaf-1 is stronger than its interaction with native exogenous Apaf-1 as indicated by dominant negative effect of ΔApaf-1 on caspase-3 processing.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Apoptosis , Apoptotic Protease-Activating Factor 1/chemistry , Biopolymers/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell-Free System , Chromatography, Affinity , Chromatography, Gel , Enzyme Activation , HEK293 Cells , Humans , Luciferases/metabolism , Protein Binding , Proteolysis , WD40 Repeats
4.
Phys Chem Chem Phys ; 20(3): 1964-1973, 2018 Jan 17.
Article in English | MEDLINE | ID: mdl-29299551

ABSTRACT

The formation of a heptameric apoptosome is a crucial event in the intrinsic cell death pathway. Considerable progress has been made towards unraveling the constituents and the structure of the apoptosome as well as the mechanism of apoptosome-mediated caspase-9 activation. However, a significant gap remains in the understanding of this process, i.e., how seven Apaf-1·cytochrome c complexes stepwisely assemble into an apoptosome. Here, we construct a biophysical model that incorporates current biochemical knowledge about the formation of apoptosome. We propose 11 elementary routes and enumerate all 2047 possible assembly pathways from the Apaf-1·cytochrome c complex to the heptameric apoptosome. By combining mathematical analysis and numerical simulation, we find that two elementary routes are the most favorable biochemical reaction routes and there are 52 optimal assembly pathways which are economical and relatively fast. Our study yields the first comprehensive analysis of apoptosome assembly and provides insights into complex assembly pathways.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Cytochromes c/metabolism , Humans , Kinetics , Models, Molecular
5.
Structure ; 25(4): 575-577, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28380338

ABSTRACT

While earlier studies of Apaf-1 holo-apoptosome architecture revealed the spectacular heptameric wheel-like structure formed by Apaf-1, the central CARD disk responsible for caspase-9 recruitment remained incompletely resolved. In a recent issue of Structure, Su et al. (2017) describe a crystal structure of the complex between Apaf-1 CARD and caspase-9 CARD. Together with two recent cryo-EM structures, this work brings us closer to a full view of the holo-apoptosome.


Subject(s)
Apoptosomes/chemistry , Caspase 9/chemistry , Apoptosis , Apoptotic Protease-Activating Factor 1/chemistry
6.
Proc Natl Acad Sci U S A ; 114(7): 1542-1547, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28143931

ABSTRACT

Mammalian intrinsic apoptosis requires activation of the initiator caspase-9, which then cleaves and activates the effector caspases to execute cell killing. The heptameric Apaf-1 apoptosome is indispensable for caspase-9 activation by together forming a holoenzyme. The molecular mechanism of caspase-9 activation remains largely enigmatic. Here, we report the cryoelectron microscopy (cryo-EM) structure of an apoptotic holoenzyme and structure-guided biochemical analyses. The caspase recruitment domains (CARDs) of Apaf-1 and caspase-9 assemble in two different ways: a 4:4 complex docks onto the central hub of the apoptosome, and a 2:1 complex binds the periphery of the central hub. The interface between the CARD complex and the central hub is required for caspase-9 activation within the holoenzyme. Unexpectedly, the CARD of free caspase-9 strongly inhibits its proteolytic activity. These structural and biochemical findings demonstrate that the apoptosome activates caspase-9 at least in part through sequestration of the inhibitory CARD domain.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Holoenzymes/metabolism , Apoptosis , Apoptosomes/chemistry , Apoptosomes/ultrastructure , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/genetics , Caspase 9/chemistry , Caspase 9/genetics , Caspase Activation and Recruitment Domain/genetics , Cryoelectron Microscopy , Enzyme Activation , Holoenzymes/chemistry , Holoenzymes/ultrastructure , Humans , Models, Molecular , Mutation , Protein Binding , Protein Domains , Protein Multimerization
7.
Cell Death Differ ; 24(4): 626-637, 2017 04.
Article in English | MEDLINE | ID: mdl-28211871

ABSTRACT

The Rb/E2F network has a critical role in regulating cell cycle progression and cell fate decisions. It is dysfunctional in virtually all human cancers, because of genetic lesions that cause overexpression of activators, inactivation of repressors, or both. Paradoxically, the downstream target of this network, E2F1, is rarely strongly overexpressed in cancer. E2F1 can induce both proliferation and apoptosis but the factors governing these critical cell fate decisions remain unclear. Previous studies have focused on qualitative mechanisms such as differential cofactors, posttranslational modification or state of other signaling pathways as modifiers of the cell fate decisions downstream of E2F1 activation. In contrast, the importance of the expression levels of E2F1 itself in dictating the downstream phenotypes has not been rigorously studied, partly due to the limited resolution of traditional population-level measurements. Here, through single-cell quantitative analysis, we demonstrate that E2F1 expression levels have a critical role in determining the fate of individual cells. Low levels of exogenous E2F1 promote proliferation, moderate levels induce G1, G2 and mitotic cell cycle arrest, and very high levels promote apoptosis. These multiple anti-proliferative mechanisms result in a strong selection pressure leading to rapid elimination of E2F1-overexpressing cells from the population. RNA-sequencing and RT-PCR revealed that low levels of E2F1 are sufficient to induce numerous cell cycle-promoting genes, intermediate levels induce growth arrest genes (i.e., p18, p19 and p27), whereas higher levels are necessary to induce key apoptotic E2F1 targets APAF1, PUMA, HRK and BIM. Finally, treatment of a lung cancer cell line with a proteasome inhibitor, MLN2238, resulted in an E2F1-dependent mitotic arrest and apoptosis, confirming the role of endogenous E2F1 levels in these phenotypes. The strong anti-proliferative activity of moderately overexpressed E2F1 in multiple cancer types suggests that targeting E2F1 for upregulation may represent an attractive therapeutic strategy in cancer.


Subject(s)
Apoptosis , E2F1 Transcription Factor/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Bcl-2-Like Protein 11/chemistry , Bcl-2-Like Protein 11/metabolism , Boron Compounds/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , E2F1 Transcription Factor/genetics , Glycine/analogs & derivatives , Glycine/pharmacology , HCT116 Cells , Histones/metabolism , Humans , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Tamoxifen/toxicity , Time-Lapse Imaging
8.
Structure ; 25(3): 407-420, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28111022

ABSTRACT

Death domain (DD)-fold assemblies play a crucial role in regulating the signaling to cell survival or death. Here we report the crystal structure of the caspase recruitment domain (CARD)-CARD disk of the human apoptosome. The structure surprisingly reveals that three 1:1 Apaf-1:procaspase-9 CARD protomers form a novel helical DD-fold assembly on the heptameric wheel-like platform of the apoptosome. The small-angle X-ray scattering and multi-angle light scattering data also support that three protomers could form an oligomeric complex similar to the crystal structure. Interestingly, the quasi-equivalent environment of CARDs could generate different quaternary CARD assemblies. We also found that the type II interaction is conserved in all DD-fold complexes, whereas the type I interaction is found only in the helical DD-fold assemblies. This study provides crucial insights into the caspase activation mechanism, which is tightly controlled by a sophisticated and highly evolved CARD assembly on the apoptosome, and also enables better understanding of the intricate DD-fold assembly.


Subject(s)
Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Apoptosis , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/chemistry , Caspase 9/chemistry , Crystallography, X-Ray , Enzyme Activation , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Multimerization , Protein Structure, Secondary , Scattering, Small Angle
9.
Sci Rep ; 6: 29820, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27443636

ABSTRACT

Apaf-1 is a central component in the apoptosis regulatory network for the treatment of apoptosis related diseases. Excessive Apaf-1 activity induced by myocardial ischemia causes cell injury. No drug targeted to Apaf-1 for treating myocardial ischemia has been reported to the best of our knowledge. In the present work, we synthesized a novel compound, ZYZ-488, which exhibited significant cardioprotective property in significantly increasing the viability of hypoxia-induced H9c2 cardiomyocytes and reducing CK and LDH leakage. Further study suggested the protective activity of ZYZ-488 dependent on its anti-apoptosis effect. This anti-apoptotic effect is most probably related to its disturbing the interaction between Apaf-1 and procaspase-9 as the target fishing and molecular docking indicated. The suppression on the activation of procaspase-9 and procaspase-3 with ZYZ-488 strongly suggested that compound ZYZ-488 could be a novel inhibitor of Apaf-1. In conclusion, ZYZ-488 as a novel small molecule competitive inhibitor of Apaf-1, with the great potential for treating cardiac ischemia.


Subject(s)
Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1/antagonists & inhibitors , Cardiotonic Agents , Myocardial Ischemia/drug therapy , Myocytes, Cardiac/metabolism , Animals , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Cardiotonic Agents/chemical synthesis , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Caspase 3/chemistry , Caspase 3/metabolism , Caspase 9/chemistry , Caspase 9/metabolism , Cell Line , Molecular Docking Simulation , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Rats
10.
Sci Rep ; 5: 15132, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26459935

ABSTRACT

The biological effects of microRNAs (miRNAs) and TNF-α in atherosclerosis have been widely studied. The circulating miR-17-92 cluster has been recently shown to be significantly downregulated in patients with injured vascular endothelium. However, it remains unclear whether the miR-17-92 cluster plays a significant role in vascular endothelial repair. The aim of this study was to investigate the relationship between the miR-17-92 cluster and TNF-α-induced endothelial cell apoptosis. We determined that the down-regulation of miR-19b level among patients with coronary artery disease was consistent with miRNA expression changes in endothelial cells following 24 h of TNF-α treatment. In vitro, the overexpression of miR-19b significantly alleviated the endothelial cells apoptosis, whereas the inhibition of miR-19b significantly enhanced apoptosis. The increased levels of Afap1 and caspase7 observed in our apoptosis model could be reduced by miR-19b, and this effect could be due to miR-19b binding 3'-UTRs of Afap1 and caspase7 mRNA. Therefore our results indicate that miR-19b plays a key role in the attenuation of TNF-α-induced endothelial cell apoptosis and that this function is closely linked to the Apaf1/caspase-dependent pathway.


Subject(s)
Apoptosis/genetics , Coronary Artery Disease/genetics , Endothelial Cells/metabolism , MicroRNAs/genetics , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/genetics , Binding Sites , Caspase 7/chemistry , Caspase 7/genetics , Coronary Artery Disease/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MicroRNAs/blood , MicroRNAs/chemistry , Multigene Family , PTEN Phosphohydrolase/genetics , RNA Interference , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Int J Biol Macromol ; 81: 370-4, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26277751

ABSTRACT

Apaf-1, the key element of apoptotic mitochondrial pathway, normally exists in an auto-inhibited form inside the cytosol. WRD-domain of Apaf-1 has a critical role in the preservation of auto-inhibited form; however the underlying mechanism is unclear. It seems the salt bridges between WRD and NOD domains are involved in maintaining the inactive conformation of Apaf-1. At the present study, we have investigated the effect of E546-R907 salt bridge on the maintenance of auto-inhibited form of human Apaf-1. E546 is mutated to glutamine (Q) and arginine (R). Over-expression of wild type Apaf-1 and its E546Q and E546R variants in HEK293T cells does not induce apoptosis unlike - HL-60 cancer cell line. In vitro apoptosome formation assay showed that all variants are cytochrome c and dATP dependent to form apoptosome and activate endogenous procaspase-9 in Apaf-1-knockout MEF cell line. These results suggest that E546 is not a critical residue for preservation of auto-inhibited Apaf-1. Furthermore, the behavior of Apaf-1 variants for in vitro apoptosome formation in HEK293T cell is similar to exogenous wild type Apaf-1. Wild type and its variants can form apoptosome in HEK293T cell with different procaspase-3 processing pattern in the presence and absence of exogenous cytochrome c and dATP.


Subject(s)
Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Arginine/chemistry , Glutamic Acid/chemistry , Animals , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Arginine/metabolism , Caspase 9/metabolism , Codon , Cytochromes c/metabolism , Deoxyadenine Nucleotides , Gene Expression , Gene Knockout Techniques , Glutamic Acid/metabolism , HEK293 Cells , Humans , Mice , Models, Molecular , Mutation , Protein Conformation
12.
Biol Direct ; 10: 29, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26014357

ABSTRACT

BACKGROUND: Binding of cytochrome c, released from the damaged mitochondria, to the apoptotic protease activating factor 1 (Apaf-1) is a key event in the apoptotic signaling cascade. The binding triggers a major domain rearrangement in Apaf-1, which leads to oligomerization of Apaf-1/cytochrome c complexes into an apoptosome. Despite the availability of crystal structures of cytochrome c and Apaf-1 and cryo-electron microscopy models of the entire apoptosome, the binding mode of cytochrome c to Apaf-1, as well as the nature of the amino acid residues of Apaf-1 involved remain obscure. RESULTS: We investigated the interaction between cytochrome c and Apaf-1 by combining several modeling approaches. We have applied protein-protein docking and energy minimization, evaluated the resulting models of the Apaf-1/cytochrome c complex, and carried out a further analysis by means of molecular dynamics simulations. We ended up with a single model structure where all the lysine residues of cytochrome c that are known as functionally-relevant were involved in forming salt bridges with acidic residues of Apaf-1. This model has revealed three distinctive bifurcated salt bridges, each involving a single lysine residue of cytochrome c and two neighboring acidic resides of Apaf-1. Salt bridge-forming amino acids of Apaf-1 showed a clear evolutionary pattern within Metazoa, with pairs of acidic residues of Apaf-1, involved in bifurcated salt bridges, reaching their highest numbers in the sequences of vertebrates, in which the cytochrome c-mediated mechanism of apoptosome formation seems to be typical. CONCLUSIONS: The reported model of an Apaf-1/cytochrome c complex provides insights in the nature of protein-protein interactions which are hard to observe in crystallographic or electron microscopy studies. Bifurcated salt bridges can be expected to be stronger than simple salt bridges, and their formation might promote the conformational change of Apaf-1, leading to the formation of an apoptosome. Combination of structural and sequence analyses provides hints on the evolution of the cytochrome c-mediated apoptosis.


Subject(s)
Apoptosomes , Apoptotic Protease-Activating Factor 1/chemistry , Cytochromes c/chemistry , Animals , Apoptosis , Horses , Humans , Hydrogen Bonding , Lysine/chemistry , Microscopy, Electron , Molecular Dynamics Simulation , Phylogeny , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Salts/chemistry , Static Electricity
13.
J Biol Chem ; 289(38): 26277-26289, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25107908

ABSTRACT

The protease caspase-9 is activated on the apoptosome, a multiprotein signal transduction platform that assembles in response to mitochondria-dependent apoptosis initiation. Despite extensive molecular research, the assembly of the holo-apoptosome and the process of caspase-9 activation remain incompletely understood. Here, we therefore integrated quantitative data on the molecular interactions and proteolytic processes during apoptosome formation and apoptosis execution and conducted mathematical simulations to investigate the resulting biochemical signaling, quantitatively and kinetically. Interestingly, when implementing the homodimerization of procaspase-9 as a prerequisite for activation, the calculated kinetics of apoptosis execution and the efficacy of caspase-3 activation failed to replicate experimental data. In contrast, assuming a scenario in which procaspase-9 is activated allosterically upon binding to the apoptosome backbone, the mathematical simulations quantitatively and kinetically reproduced all experimental data. These data included a XIAP threshold concentration at which apoptosis execution is suppressed in HeLa cervical cancer cells, half-times of procaspase-9 processing, as well as the molecular timer function of the apoptosome. Our study therefore provides novel mechanistic insight into apoptosome-dependent apoptosis execution and suggests that caspase-9 is activated allosterically by binding to the apoptosome backbone. Our findings challenge the currently prevailing dogma that all initiator procaspases require homodimerization for activation.


Subject(s)
Apoptosis , Apoptosomes/physiology , Caspase 9/physiology , Protein Precursors/physiology , Allosteric Regulation , Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/physiology , Caspase 9/chemistry , Catalytic Domain , Computer Simulation , Enzyme Activation , HeLa Cells , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Multimerization , Protein Precursors/chemistry , Systems Biology
14.
BMC Cancer ; 14: 165, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24606760

ABSTRACT

BACKGROUND: Translationally controlled tumor protein (TCTP), alternatively called fortilin, is believed to be involved in the development of the chemoresistance of tumor cells against anticancer drugs such as etoposide, taxol, and oxaliplatin, the underlying mechanisms of which still remain elusive. METHODS: Cell death analysis of TCTP-overexpressing HeLa cells was performed following etoposide treatment to assess the mitochondria-dependent apoptosis. Apoptotic pathway was analyzed through measuring the cleavage of epidermal growth factor receptor (EGFR) and phospholipase C-γ (PLC-γ), caspase activation, mitochondrial membrane perturbation, and cytochrome c release by flow cytometry and western blotting. To clarify the role of TCTP in the inhibition of apoptosome, in vitro apoptosome reconstitution and immunoprecipitation was used. Pull-down assay and silver staining using the variants of Apaf-1 protein was applied to identify the domain that is responsible for its interaction with TCTP. RESULTS: In the present study, we confirmed that adenoviral overexpression of TCTP protects HeLa cells from cell death induced by cytotoxic drugs such as taxol and etoposide. TCTP antagonized the mitochondria-dependent apoptotic pathway following etoposide treatment, including mitochondrial membrane damage and resultant cytochrome c release, activation of caspase-9, and -3, and eventually, the cleavage of EGFR and PLC-γ. More importantly, TCTP interacts with the caspase recruitment domain (CARD) of Apaf-1 and is incorporated into the heptameric Apaf-1 complex, and that C-terminal cleaved TCTP specifically associates with Apaf-1 of apoptosome in apoptosome-forming condition thereby inhibiting the amplification of caspase cascade. CONCLUSIONS: TCTP protects the cancer cells from etoposide-induced cell death by inhibiting the mitochondria-mediated apoptotic pathway. Interaction of TCTP with Apaf-1 in apoptosome is involved in the molecular mechanism of TCTP-induced chemoresistance. These findings suggest that TCTP may serve as a therapeutic target for chemoresistance in cancer treatment.


Subject(s)
Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Epistasis, Genetic , Apoptosis/drug effects , Apoptosis/genetics , Apoptotic Protease-Activating Factor 1/chemistry , Caspase 3/metabolism , Caspase 9/metabolism , Cell Death/drug effects , Cell Death/genetics , Cytochromes c/metabolism , DNA Fragmentation , ErbB Receptors/genetics , Etoposide/pharmacology , Gene Expression , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Phospholipase C gamma/genetics , Protein Binding , Protein Interaction Domains and Motifs , Topoisomerase II Inhibitors/pharmacology , Tumor Protein, Translationally-Controlled 1
15.
Biochim Biophys Acta ; 1843(4): 685-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424093

ABSTRACT

Apoptosis is essential in the death process induced by Amyloid-ß (Aß), a major constituent of diffuse plaques found in Alzheimer's disease patients. However, we have found that caspase activation and cell death induced by staurosporine, employed to induce the intrinsic mitochondria-dependent apoptotic pathway, were significantly reduced by 42 amino-acid Aß42, implying that the peptide also has a negative effect on the apoptotic process. The inhibitory effect of Aß42 on the apoptotic pathway is associated with its interaction with procaspase-9 and consequent inhibition of Apaf-1 apoptosome assembly. We detected the inhibitory effect in the early stage (<8h) of apoptosis, but later caspase activation becomes obvious. Thus we inferred that the inhibitory process on apoptosis begins at an early stage, and the later robust activation surpasses it. We propose that the apoptotic manifestation in Aß-treated cells is a combined consequence of those anti- and pro-apoptotic processes.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apoptosis/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Apoptosis/drug effects , Apoptosomes/chemistry , Apoptosomes/ultrastructure , Apoptotic Protease-Activating Factor 1/chemistry , Caspase 3/metabolism , Cell-Free System , HeLa Cells , Humans , Protein Binding , Signal Transduction/drug effects , Staurosporine/pharmacology
16.
Curr Cancer Drug Targets ; 14(1): 2-29, 2014.
Article in English | MEDLINE | ID: mdl-24200083

ABSTRACT

Understanding the molecular mechanisms and the signaling pathways that underlie the pathology of cancer progression is crucial for the development of novel diagnostic and therapeutic tools. A major common mechanism used by cells to regulate intracellular signal transduction pathways is reversible protein phosphorylation which results in profound changes in cellular responses. This mechanism relies on the coordinated action of two families of proteins: protein kinases and protein phosphatases. Interestingly, there are 3 to 5 times fewer phosphatases than kinases, suggesting that the specificity of substrates is not only due to the variety of the catalytic subunits but also to the diversity of the regulatory subunits. This is particularly true for PhosphoProtein Phosphatase 1 (PPP1) for which more than 200 PPP1 Interacting Proteins (PIPs) have thus far been identified. PIPs can act as targeting subunits, substrates and activity regulators. Many PPP1/PIPs complexes are involved in signaling pathways that regulate cellular growth, cell cycle and apoptosis; processes known to be deregulated in cancer. This review will describe the cellular pathways, many of which involve PPP1/PIP complexes, that when deregulated lead to cancer. Furthermore, the possibility of PPP1/PIP complexes being considered novel targets to cancer diagnostic and therapy will be addressed.


Subject(s)
Carcinogenesis/metabolism , Protein Phosphatase 1/metabolism , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , BRCA1 Protein/metabolism , Binding Sites , Caspases/metabolism , Catalytic Domain , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
17.
Structure ; 21(4): 501-15, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23561633

ABSTRACT

Apaf-1-like molecules assemble into a ring-like platform known as the apoptosome. This cell death platform then activates procaspases in the intrinsic cell death pathway. In this review, crystal structures of Apaf-1 monomers and CED-4 dimers have been combined with apoptosome structures to provide insights into the assembly of cell death platforms in humans, nematodes, and flies. In humans, the caspase recognition domains (CARDs) of procaspase-9 and Apaf-1 interact with each other to form a CARD-CARD disk, which interacts with the platform to create an asymmetric proteolysis machine. The disk tethers multiple pc-9 catalytic domains to the platform to raise their local concentration, and this leads to zymogen activation. These findings have now set the stage for further studies of this critical activation process on the apoptosome.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis/physiology , Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Calcium-Binding Proteins/chemistry , Caspase 9/metabolism , Models, Molecular , Animals , Apoptosis Regulatory Proteins/metabolism , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Caenorhabditis elegans , Calcium-Binding Proteins/metabolism , Dimerization , Drosophila , Enzyme Activation/physiology , Humans , Protein Conformation , Species Specificity
18.
Biochemistry (Mosc) ; 78(2): 157-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23581986

ABSTRACT

We have earlier shown that the 5'-untranslated region (5' UTR) of the mRNA coding for activation factor of apoptotic peptidase 1 (Apaf-1) can direct translation in vivo by strictly 5' end-dependent way even in the absence of m(7)G-cap. Dependence of translational efficiency on the cap availability for this mRNA turned out to be relatively low. In this study we demonstrate that this surprising phenomenon is determined the 5'-proximal part (domains I and II) of highly structured Apaf-1 5' UTR. Remarkably, domain II by itself was able to reduce dependence of the mRNA on the cap on its transferring to a short 5' UTR derived from a standard vector. We suggest that the low cap-dependence inherent to some cellular mRNAs may have an important physiological significance under those stress conditions when the function of cap-binding factor eIF4E is impaired.


Subject(s)
5' Untranslated Regions , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/genetics , Protein Biosynthesis , RNA, Messenger/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Base Sequence , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Protein Structure, Secondary , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/chemistry , RNA, Messenger/genetics
19.
J Biol Chem ; 288(21): 15142-7, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23572523

ABSTRACT

Maturation of the single-chain caspase-9 zymogen through autoproteolytic processing is mediated by the Apaf-1 apoptosome at the onset of apoptosis. Processed caspase-9 and the apoptosome form a holoenzyme with robust proteolytic activity that is 2-3 orders of magnitude higher than that of free processed caspase-9. An unresolved important question is the role of caspase-9 processing, with some experimental data suggesting its dispensability. In this study, we demonstrate that, in contrast to wild-type caspase-9, the unprocessed single-chain caspase-9 triple mutant E306A/D315A/D330A (Casp9-TM) could no longer be adequately activated by the apoptosome. Compared with the protease activity of wild-type caspase-9, that of Casp9-TM in the presence of the apoptosome was drastically reduced. The crippled protease activity of Casp9-TM in the presence of the apoptosome is likely attributable to a markedly reduced ability of Casp9-TM to form homodimers. These data identify an essential role for the autoproteolytic processing of caspase-9 in its activation.


Subject(s)
Apoptosomes/chemistry , Apoptotic Protease-Activating Factor 1/chemistry , Caspase 9/chemistry , Enzyme Precursors/chemistry , Protein Multimerization/physiology , Proteolysis , Amino Acid Substitution , Apoptosomes/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Enzyme Activation/physiology , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Humans , Mutation, Missense
20.
Biochemistry ; 52(13): 2319-27, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23521171

ABSTRACT

Apoptosome assembly is highly regulated in the intrinsic cell death pathway. To better understand this step, we created an improved model of the human apoptosome using a crystal structure of full length Apaf-1 and a single particle, electron density map at ~9.5 Å resolution. The apoptosome model includes N-terminal domains of Apaf-1, cognate ß-propellers, and cytochrome c. A direct comparison of Apaf-1 in the apoptosome and as a monomer reveals conformational changes that occur during the first two steps of assembly. This includes an induced-fit mechanism for cytochrome c binding to regulatory ß-propellers, which is dependent on shape and charge complementarity, and a large rotation of the nucleotide binding module during nucleotide exchange. These linked conformational changes create an extended Apaf-1 monomer and drive apoptosome assembly. Moreover, the N-terminal CARD in the inactive Apaf-1 monomer is not shielded from other proteins by ß-propellers. Hence, the Apaf-1 CARD may be free to interact with a procaspase-9 CARD either before or during apoptosome assembly. Irrespective of the timing, the end product of assembly is a holo-apoptosome with an acentric CARD-CARD disk and tethered pc-9 catalytic domains. Subsequent activation of pc-9 leads to a proteolytic cascade and cell death.


Subject(s)
Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/chemistry , Apoptotic Protease-Activating Factor 1/metabolism , Apoptosomes/chemistry , Cytochromes c/chemistry , Cytochromes c/metabolism , Humans , Models, Molecular , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...