Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
1.
Proc Natl Acad Sci U S A ; 121(9): e2312784121, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38381783

ABSTRACT

The onset of apoptosis is characterized by a cascade of caspase activation, where initiator caspases are activated by a multimeric adaptor complex known as the apoptosome. In Drosophila melanogaster, the initiator caspase Dronc undergoes autocatalytic activation in the presence of the Dark apoptosome. Despite rigorous investigations, the activation mechanism for Dronc remains elusive. Here, we report the cryo-EM structures of an auto-inhibited Dark monomer and a single-layered, multimeric Dark/Dronc complex. Our biochemical analysis suggests that the auto-inhibited Dark oligomerizes upon binding to Dronc, which is sufficient for the activation of both Dark and Dronc. In contrast, the previously observed double-ring Dark apoptosome may represent a non-functional or "off-pathway" conformation. These findings expand our understanding on the molecular mechanism of apoptosis in Drosophila.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Apoptosomes/chemistry , Caspases/metabolism , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 120(51): e2310944120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38085782

ABSTRACT

Mitochondrial apoptotic signaling cascades lead to the formation of the apoptosome, a 1.1-MDa heptameric protein scaffold that recruits and activates the caspase-9 protease. Once activated, caspase-9 cleaves and activates downstream effector caspases, triggering the onset of cell death through caspase-mediated proteolysis of cellular proteins. Failure to activate caspase-9 enables the evasion of programmed cell death, which occurs in various forms of cancer. Despite the critical apoptotic function of caspase-9, the structural mechanism by which it is activated on the apoptosome has remained elusive. Here, we used a combination of methyl-transverse relaxation-optimized NMR spectroscopy, protein engineering, and biochemical assays to study the activation of caspase-9 bound to the apoptosome. In the absence of peptide substrate, we observed that both caspase-9 and its isolated protease domain (PD) only very weakly dimerize with dissociation constants in the millimolar range. Methyl-NMR spectra of isotope-labeled caspase-9, within the 1.3-MDa native apoptosome complex or an engineered 480-kDa apoptosome mimic, reveal that the caspase-9 PD remains monomeric after recruitment to the scaffold. Binding to the apoptosome, therefore, organizes caspase-9 PDs so that they can rapidly and extensively dimerize only when substrate is present, providing an important layer in the regulation of caspase-9 activation. Our work highlights the unique role of NMR spectroscopy to structurally characterize protein domains that are flexibly tethered to large scaffolds, even in cases where the molecular targets are in excess of 1 MDa, as in the present example.


Subject(s)
Apoptosomes , Caspases , Caspase 9/metabolism , Apoptosomes/chemistry , Caspases/metabolism , Apoptosis , Magnetic Resonance Spectroscopy , Caspase 3/metabolism
3.
Life Sci Alliance ; 6(9)2023 09.
Article in English | MEDLINE | ID: mdl-37402593

ABSTRACT

In Caenorhabditis elegans (C. elegans), onset of programmed cell death is marked with the activation of CED-3, a process that requires assembly of the CED-4 apoptosome. Activated CED-3 forms a holoenzyme with the CED-4 apoptosome to cleave a wide range of substrates, leading to irreversible cell death. Despite decades of investigations, the underlying mechanism of CED-4-facilitated CED-3 activation remains elusive. Here, we report cryo-EM structures of the CED-4 apoptosome and three distinct CED-4/CED-3 complexes that mimic different activation stages for CED-3. In addition to the previously reported octamer in crystal structures, CED-4, alone or in complex with CED-3, exists in multiple oligomeric states. Supported by biochemical analyses, we show that the conserved CARD-CARD interaction promotes CED-3 activation, and initiation of programmed cell death is regulated by the dynamic organization of the CED-4 apoptosome.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Apoptosomes/metabolism , Apoptosis
4.
Mol Biol Cell ; 34(5): ar41, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36920061

ABSTRACT

The actin cytoskeleton is a ubiquitous participant in cellular functions that maintain viability, but how it controls programmed cell death is not well understood. Here we show that in response to DNA damage, human cells form a juxtanuclear F-actin-rich territory that coordinates the organized progression of apoptosome assembly to caspase activation. This cytoskeletal compartment is created by the actin nucleation factors JMY, WHAMM, and the Arp2/3 complex, and it excludes proteins that inhibit JMY and WHAMM activity. Within the territory, mitochondria undergo outer membrane permeabilization and JMY localization overlaps with punctate structures containing the core apoptosome components cytochrome c and Apaf-1. The F-actin-rich area also encompasses initiator caspase-9 and clusters of a cleaved form of executioner caspase-3 but restricts accessibility of the caspase inhibitor XIAP. The clustering and potency of caspase-3 activation are positively regulated by the amount of actin polymerized by JMY and WHAMM. These results indicate that JMY-mediated actin reorganization functions in apoptotic signaling by coupling the biogenesis of apoptosomes to the localized processing of caspases.


Subject(s)
Actins , Apoptosomes , Humans , Actins/metabolism , Caspase 3 , Apoptosomes/metabolism , Apoptosis/physiology , Caspases/metabolism , Actin Cytoskeleton/metabolism , DNA Damage , Membrane Proteins/metabolism , Microtubule-Associated Proteins
5.
Dis Markers ; 2022: 8809956, 2022.
Article in English | MEDLINE | ID: mdl-36225197

ABSTRACT

Lung adenocarcinoma (LUAD) is the most common subtype of nonsmall cell lung cancer. Cytochrome c (Cyt c), which is produced from mitochondria, interacts with a protein called Apaf-1 to form the heptameric apoptosome. This heptameric apoptosome then activates the caspase cascade, which ultimately results in the execution of apoptosis. The purpose of our research was to discover a new prognostic model that is based on cytochrome c-related genes (CCRGs) for LUAD patients. Through LASSO regression analysis conducted on the LUAD datasets included in the TCGA datasets, a CCRGs signature was created. The diagnostic accuracy of the multigene signature was verified by an independent source using the GSE31210 and GSE72094 datasets. The GO and KEGG enrichment analysis were performed. In this study, there were 159 differentially expressed CCRGs in the TCGA dataset, while there were 68 differentially expressed CCRGs in the GSE31210 dataset. Additionally, there were 57 genes that overlapped across the two datasets. Using LASSO and Cox regression analysis, a signature consisting of 12 differentially expressed CCRGs was developed from the total of 57 such genes. On the basis of their risk ratings, patients were categorized into high-risk and low-risk categories, with low-risk patients having lower risk scores and a greater likelihood of surviving the disease. Univariate and multivariate analyses both concluded that this signature is an independent risk factor for LUAD. ROC curves demonstrated that this risk signature is capable of accurately predicting the 1-year, 2-year, 3-year, and 5-year survival rates of patients who have LUAD. The infiltration of antigen-presenting cells was higher in the low-risk group, such as aDCs, DCs, pDCs, and iDCs. The expression of multiple immune checkpoints was significantly higher in the low-risk group, such as BTLA, CD28, and CD86. Finally, we showed that the signature can be used to predict the drug sensitivity of already available or under investigational drugs. Overall, patient classification and individualized therapy options may benefit from this study's development of a powerful gene signature with high value for prognostic prediction in LUAD.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adenocarcinoma of Lung/pathology , Apoptosomes , CD28 Antigens , Caspases , Cytochromes c , Drugs, Investigational , Humans , Lung Neoplasms/pathology , Prognosis
6.
Adv Sci (Weinh) ; 9(28): e2201889, 2022 10.
Article in English | MEDLINE | ID: mdl-35975461

ABSTRACT

Chemotherapeutics remain the first choice for advanced gastric cancers (GCs). However, drug resistance and unavoidable severe toxicity lead to chemotherapy failure and poor prognosis. Long noncoding RNAs (lncRNAs) play critical roles in tumor progression in many cancers, including GC. Here, through RNA screening, an apoptotic protease-activating factor 1 (APAF1)-binding lncRNA (ABL) that is significantly elevated in cancerous GC tissues and an independent prognostic factor for GC patients is identified. Moreover, ABL overexpression inhibits GC cell apoptosis and promotes GC cell survival and multidrug resistance in GC xenograft and organoid models. Mechanistically, ABL directly binds to the RNA-binding protein IGF2BP1 via its KH1/2 domain, and then IGF2BP1 further recognizes the METTL3-mediated m6A modification on ABL, which maintains ABL stability. In addition, ABL can bind to the WD1/WD2 domain of APAF1, which competitively prevent cytochrome c from interacting with APAF1, blocking apoptosome assembly and caspase-9/3 activation; these events lead to resistance to cell death in GC cells. Intriguingly, targeting ABL using encapsulated liposomal siRNA can significantly enhance the sensitivity of GC cells to chemotherapy. Collectively, the results suggest that ABL can be a potential prognostic biomarker and therapeutic target in GC.


Subject(s)
RNA, Long Noncoding , Stomach Neoplasms , Apoptosis/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Biomarkers , Caspase 9/metabolism , Cytochromes c/metabolism , Cytochromes c/therapeutic use , Drug Resistance, Multiple , Humans , Methyltransferases/metabolism , Methyltransferases/therapeutic use , RNA, Long Noncoding/genetics , RNA, Small Interfering/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
7.
Environ Toxicol Pharmacol ; 95: 103964, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36028164

ABSTRACT

Cytotoxic drugs have been recognized by the European Union as the potential threat in the aquatic environment. As a typical cytotoxic drug, effects of long-term exposure to cisplatin at the environmentally relevant concentrations on the crustacean health and its molecular mechanism remain undetermined. In this study, the growth and reproduction of Daphnia magna resulting from cisplatin exposure were initially assessed. While the phenotypes were not altered in 2 µg L-1, 20 µg L-1, and 200 µg L-1 treatment groups, cisplatin at 500 µg L-1 significantly reduced the offspring number to 8-13 neonates in each brood, which was lower than 13-27 neonates in the control group. In addition to the delay in the time of first pregnancy, the body length was decreased by approximate 12.13% at day 7. Meanwhile, all daphnids died after exposure to 500 µg L-1 cisplatin for 17 days. Transcriptome profiling bioassays were performed for 10 days to explore the alternation at the molecular level. Briefly, 980 (257 up- and 723 down-regulated), 429 (182 up- and 247 down-regulated) and 1984 (616 up-regulated and 1368 down-regulated) genes were differentially expressed (adj p < 0.05) in low (2 µg L-1), medium (200 µg L-1) and high (500 µg L-1) cisplatin treatment groups, respectively. Differentially expressed genes were primarily enriched in the digestion and absorption, nerve conduction, endocrine interference, and circulatory related pathways. Specifically, the down-regulated digestive secretion and nutrient absorption and neuronal conduction pathways may lead to insufficient energy supply involved in growth and reproduction, and hinder ovarian development and cell growth. Down-regulation of ovarian steroids and relaxin signaling pathways may be related to the reduction of offspring number and delayed pregnancy, and reduced body length of D. magna may attribute to the enrichment of insulin secretion pathway. In addition, the death of D. magna may result from the reduced expression of genes in cardiomyocyte contraction and apoptosome processes. Taken together, this study revealed the potential toxic mechanism of cisplatin in a model water flea.


Subject(s)
Antineoplastic Agents , Cladocera , Insulins , Relaxin , Water Pollutants, Chemical , Animals , Antineoplastic Agents/toxicity , Apoptosomes , Cisplatin/toxicity , Daphnia/genetics , Insulins/pharmacology , Relaxin/pharmacology , Reproduction , Transcriptome , Water Pollutants, Chemical/toxicity
8.
Biochem J ; 479(3): 357-384, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35147165

ABSTRACT

Regulated cell death is a vital and dynamic process in multicellular organisms that maintains tissue homeostasis and eliminates potentially dangerous cells. Apoptosis, one of the better-known forms of regulated cell death, is activated when cell-surface death receptors like Fas are engaged by their ligands (the extrinsic pathway) or when BCL-2-family pro-apoptotic proteins cause the permeabilization of the mitochondrial outer membrane (the intrinsic pathway). Both the intrinsic and extrinsic pathways of apoptosis lead to the activation of a family of proteases, the caspases, which are responsible for the final cell demise in the so-called execution phase of apoptosis. In this review, I will first discuss the most common types of regulated cell death on a morphological basis. I will then consider in detail the molecular pathways of intrinsic and extrinsic apoptosis, discussing how they are activated in response to specific stimuli and are sometimes overlapping. In-depth knowledge of the cellular mechanisms of apoptosis is becoming more and more important not only in the field of cellular and molecular biology but also for its translational potential in several pathologies, including neurodegeneration and cancer.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , Animals , Apoptosomes/physiology , Apoptosomes/ultrastructure , Autophagy , Caspases/physiology , Humans , Invertebrates/cytology , Ligands , Lysosomes/physiology , Macrophages/physiology , Mitochondrial Membranes/physiology , Necrosis , Neoplasm Proteins/physiology , Permeability , Phagocytosis , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Death Domain/physiology
9.
J Mol Biol ; 434(4): 167333, 2022 02 28.
Article in English | MEDLINE | ID: mdl-34756921

ABSTRACT

Pyroptosis has been described in mammalian systems to be a form of programmed cell death that is important in immune function through the subsequent release of cytokines and immune effectors upon cell bursting. This form of cell death has been increasingly well-characterized in mammals and can occur using alternative routes however, across phyla, there has been little evidence for the existence of pyroptosis. Here we provide evidence for an ancient origin of pyroptosis in an in vivo immune scenario in Drosophila melanogaster. Crystal cells, a type of insect blood cell, were recruited to wounds and ruptured subsequently releasing their cytosolic content in a caspase-dependent manner. This inflammatory-based programmed cell death mechanism fits the features of pyroptosis, never before described in an in vivo immune scenario in insects and relies on ancient apoptotic machinery to induce proto-pyroptosis. Further, we unveil key players upstream in the activation of cell death in these cells including the apoptosome which may play an alternative role akin to the inflammasome in proto-pyroptosis. Thus, Drosophila may be a suitable model for studying the functional significance of pyroptosis in the innate immune system.


Subject(s)
Apoptosomes , Drosophila melanogaster , Inflammasomes , Pyroptosis , Animals , Caspases/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Mammals
10.
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
11.
Biochem J ; 478(3): 669-684, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33480393

ABSTRACT

Mutation of cytochrome c in humans causes mild autosomal dominant thrombocytopenia. The role of cytochrome c in platelet formation, and the molecular mechanism underlying the association of cytochrome c mutations with thrombocytopenia remains unknown, although a gain-of-function is most likely. Cytochrome c contributes to several cellular processes, with an exchange between conformational states proposed to regulate changes in function. Here, we use experimental and computational approaches to determine whether pathogenic variants share changes in structure and function, and to understand how these changes might occur. Three pathogenic variants (G41S, Y48H, A51V) cause an increase in apoptosome activation and peroxidase activity. Molecular dynamics simulations of these variants, and two non-naturally occurring variants (G41A, G41T), indicate that increased apoptosome activation correlates with the increased overall flexibility of cytochrome c, particularly movement of the Ω loops. Crystal structures of Y48H and G41T complement these studies which overall suggest that the binding of cytochrome c to apoptotic protease activating factor-1 (Apaf-1) may involve an 'induced fit' mechanism which is enhanced in the more conformationally mobile variants. In contrast, peroxidase activity did not significantly correlate with protein dynamics. Thus, the mechanism by which the variants increase peroxidase activity is not related to the conformational dynamics of the native hexacoordinate state of cytochrome c. Recent molecular dynamics data proposing conformational mobility of specific cytochrome c regions underpins changes in reduction potential and alkaline transition pK was not fully supported. These data highlight that conformational dynamics of cytochrome c drive some but not all of its properties and activities.


Subject(s)
Apoptosis/physiology , Cytochromes c/chemistry , Mutation, Missense , Point Mutation , Amino Acid Substitution , Apoptosomes , Crystallography, X-Ray , Cytochromes c/genetics , Cytochromes c/isolation & purification , Cytochromes c/metabolism , Humans , Hydrogen Bonding , Hydrogen Peroxide/metabolism , Hydrogen-Ion Concentration , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship , U937 Cells
12.
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
13.
Development ; 147(14)2020 07 24.
Article in English | MEDLINE | ID: mdl-32709690

ABSTRACT

Cell death is an important facet of animal development. In some developing tissues, death is the ultimate fate of over 80% of generated cells. Although recent studies have delineated a bewildering number of cell death mechanisms, most have only been observed in pathological contexts, and only a small number drive normal development. This Primer outlines the important roles, different types and molecular players regulating developmental cell death, and discusses recent findings with which the field currently grapples. We also clarify terminology, to distinguish between developmental cell death mechanisms, for which there is evidence for evolutionary selection, and cell death that follows genetic, chemical or physical injury. Finally, we suggest how advances in understanding developmental cell death may provide insights into the molecular basis of developmental abnormalities and pathological cell death in disease.


Subject(s)
Cell Death , Invertebrates/growth & development , Mammals/growth & development , Animals , Apoptosis/genetics , Apoptosomes/metabolism , Autophagy/genetics , Caspases/metabolism , Cell Death/genetics , Gene Expression Regulation, Developmental , Invertebrates/metabolism , Mammals/metabolism , Signal Transduction
14.
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
15.
J Cell Sci ; 133(10)2020 05 27.
Article in English | MEDLINE | ID: mdl-32461338

ABSTRACT

Detection of the apoptosis signature becomes central in understanding cell death modes. We present here a whole-cell biosensor that detects Apaf-1 association and apoptosome formation using a split-luciferase complementary assay. Fusion of N-terminal (Nluc) and C-terminal (Cluc)-fragments of firefly luciferase to the N-terminus of human Apaf-1 was performed in HEK293 cells by using CRISPR-Cas9 technology. This resulted in a luminescent form of the apoptosome that we named 'Lumiptosome'. During Apaf-1 gene editing, a high number of knock-in events were observed without selection, suggesting that the Apaf-1 locus is important for the integration of exogenous transgenes. Since activation of caspase-9 is directly dependent on the apoptosome formation, measured reconstitution of luciferase activity should result from the cooperative association of Nluc-Apaf-1 and Cluc-Apaf-1. Time-response measurements also confirmed that formation of the apoptosome occurs prior to activation of caspase-3. Additionally, overexpression of the Bcl2 apoptosis regulator in transgenic and normal HEK293 cells confirmed that formation of the Lumiptosome depends on release of cytochrome c Thus, HEK293 cells that stably express the Lumiptosome can be utilized to screen pro- and anti-apoptotic drugs, and to examine Apaf-1-dependent cellular pathways.


Subject(s)
Apoptosis , Apoptosomes , Apoptosis/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Death , Cytochromes c/genetics , Cytochromes c/metabolism , HEK293 Cells , Humans
16.
Sensors (Basel) ; 20(6)2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32210205

ABSTRACT

The apoptotic protease-activating factor 1 (Apaf-1) split luciferase biosensor has been used as a biological tool for the detection of early stage of apoptosis. The effect of doxorubicin in a cell-based assay and the addition of cytochrome c and ATP in a cell-free system have been used to test the functionality of the reporter for the detection of apoptosome formation. Here, our data established a drug- and cytochrome c/ATP-independent way of apoptosis induction relying on the expression of the biosensor itself to induce formation of apoptosome. Overexpression of Apaf-1 constructs led to increased split luciferase activity and caspase-3 activity in the absence of any drug treatment. Caspase-3 activity was significantly inhibited when caspase-9DN was co-overexpressed, while the activity of the Apaf1 biosensor was significantly increased. Our results show that the Apaf-1 biosensor does not detect etoposide-induced apoptosis.


Subject(s)
Apoptosomes/metabolism , Biosensing Techniques/methods , Apoptosis/drug effects , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Enzyme Activation/drug effects , Etoposide/pharmacology , HEK293 Cells , Humans , Luciferases/metabolism
17.
Biochem Biophys Res Commun ; 523(3): 567-572, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31937411

ABSTRACT

Neuroglobin is a heme protein present in the nervous system cells of mammals and other organisms. Although cytoprotective effects of neuroglobin on neuronal damage have been reported, the physiological mechanisms of neuroglobin function remain unknown. In recent years, a role for neuroglobin as a reductant for extramitochondrial cytochrome c has been proposed. According to this hypothesis, cytoplasmic neuroglobin can interact with cytochrome c released from the mitochondria and reduce its heme group to the ferrous state, thus preventing cytochrome c-dependent assembly of the apoptosome. The interaction of neuroglobin and cytochrome c has been studied by surface plasmon resonance techniques and molecular dynamics, however the empirical evidence on the specific residues of neuroglobin and cytochrome c involved in the interaction is scarce and indirect. This study analyzes the role of five negatively charged residues in the neuroglobin surface putatively involved in the interaction with cytochrome c - Glu60, Asp63, Asp73, Glu 87 and Glu151 - by site-directed mutagenesis. Characterization of the electron transfer between neuroglobin mutants and cytochrome c indicates that Asp73 is critical for the interaction, and Glu60, Asp63 and Glu87 also contribute to the neuroglobin-cytochrome c interaction. Based on the results, structures and binding surfaces for the neuroglobin-cytochrome c complex compatible with the experimental observations are proposed. These data can guide further studies on neuroglobin function and its involvement in cytochrome c signaling cascades.


Subject(s)
Cytochromes c/metabolism , Neuroglobin/metabolism , Animals , Apoptosis , Apoptosomes/metabolism , Binding Sites , Cytochromes c/chemistry , Electron Transport , Horses , Molecular Docking Simulation , Neuroglobin/chemistry , Protein Binding , Protein Interaction Maps , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Static Electricity
18.
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
19.
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
20.
Biosci Rep ; 39(5)2019 05 31.
Article in English | MEDLINE | ID: mdl-30918105

ABSTRACT

Human breast cancer is a malignant form of tumor with a relatively high mortality rate. Although esophageal cancer-related gene 4 (ECRG4) is thought to be a possible potent tumor suppressor gene that acts to suppress breast cancer, its precise role in this disease is not understood. Herein, we assess the correlation between ECRG4 expression and DNA methylation, probing the potential epigenetic regulation of ECRG4 in breast cancer. We analyzed ECRG4 promoter methylation via methylation-specific PCR (MSPCR), bisulfite sequencing, and a promoter reporter assay in human breast cancer cell lines and samples. Gene expression was assessed by quantitative real-time PCR (qPCR), while protein levels were assessed by Western blotting. CCK8 assays were used to quantify cell growth; Esophageal cancer-related gene 4 wound healing assays were used to assess cellular migration, while flow cytometry was used to assess apoptosis and cell cycle progression. Apoptosome formation was validated via CO-IP and Western blotting. We found that human breast cancer samples exhibited increased methylation of the ECRG4 promoter and decreased ECRG4 expression. Remarkably, the down-regulation of ECRG4 was highly associated with promoter methylation, and its expression could be re-activated via 5-aza-2'-deoxycytidine treatment to induce demethylation. ECRG4 overexpression impaired breast cancer cell proliferation and migration, and led to G0/G1 cell cycle phase arrest. Moreover, ECRG4 induced the formation of the Cytc/Apaf-1/caspase-9 apoptosome and promoted breast cancer cell apoptosis. ECRG4 is silenced in human breast cancer cells and cell lines, likely owing to promoter hypermethylation. ECRG4 may act as a tumor suppressor, inhibiting proliferation and migration, inducing G0/G1 phase arrest and apoptosis via the mitochondrial apoptotic pathway.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosomes/genetics , Apoptosomes/metabolism , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Decitabine/pharmacology , Epigenesis, Genetic , Female , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Humans , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...