RESUMEN
The CED-4 homo-oligomer or apoptosome is required for initiation of programmed cell death in Caenorhabditis elegans by facilitating autocatalytic activation of the CED-3 caspase zymogen. How the CED-4 apoptosome assembles and activates CED-3 remains enigmatic. Here we report the crystal structure of the complete CED-4 apoptosome and show that it consists of eight CED-4 molecules, organized as a tetramer of an asymmetric dimer via a previously unreported interface among AAA(+) ATPases. These eight CED-4 molecules form a funnel-shaped structure. The mature CED-3 protease is monomeric in solution and forms an active holoenzyme with the CED-4 apoptosome, within which the protease activity of CED-3 is markedly stimulated. Unexpectedly, the octameric CED-4 apoptosome appears to bind only two, not eight, molecules of mature CED-3. The structure of the CED-4 apoptosome reveals shared principles for the NB-ARC family of AAA(+) ATPases and suggests a mechanism for the activation of CED-3.
Asunto(s)
Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/metabolismo , Proteínas de Unión al Calcio/química , Secuencia de Aminoácidos , Animales , Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caenorhabditis elegans/química , Caspasas/química , Cristalografía por Rayos X , Modelos Moleculares , Alineación de Secuencia , Difracción de Rayos XRESUMEN
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.
Asunto(s)
Muerte Celular , Invertebrados/crecimiento & desarrollo , Mamíferos/crecimiento & desarrollo , Animales , Apoptosis/genética , Apoptosomas/metabolismo , Autofagia/genética , Caspasas/metabolismo , Muerte Celular/genética , Regulación del Desarrollo de la Expresión Génica , Invertebrados/metabolismo , Mamíferos/metabolismo , Transducción de SeñalRESUMEN
Apoptosis is executed by a cascade of caspase activation. The autocatalytic activation of an initiator caspase, exemplified by caspase-9 in mammals or its ortholog, Dronc, in fruit flies, is facilitated by a multimeric adaptor complex known as the apoptosome. The underlying mechanism by which caspase-9 or Dronc is activated by the apoptosome remains unknown. Here we report the electron cryomicroscopic (cryo-EM) structure of the intact apoptosome from Drosophila melanogaster at 4.0 Å resolution. Analysis of the Drosophila apoptosome, which comprises 16 molecules of the Dark protein (Apaf-1 ortholog), reveals molecular determinants that support the assembly of the 2.5-MDa complex. In the absence of dATP or ATP, Dronc zymogen potently induces formation of the Dark apoptosome, within which Dronc is efficiently activated. At 4.1 Å resolution, the cryo-EM structure of the Dark apoptosome bound to the caspase recruitment domain (CARD) of Dronc (Dronc-CARD) reveals two stacked rings of Dronc-CARD that are sandwiched between two octameric rings of the Dark protein. The specific interactions between Dronc-CARD and both the CARD and the WD40 repeats of a nearby Dark protomer are indispensable for Dronc activation. These findings reveal important mechanistic insights into the activation of initiator caspase by the apoptosome.
Asunto(s)
Apoptosomas/química , Caspasas/metabolismo , Drosophila/enzimología , Modelos Moleculares , Animales , Apoptosomas/metabolismo , Proteínas de Drosophila/metabolismo , Activación Enzimática , Unión Proteica , Estructura Terciaria de ProteínaRESUMEN
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.
Asunto(s)
Apoptosis , Apoptosomas , Apoptosis/genética , Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/genética , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Muerte Celular , Citocromos c/genética , Citocromos c/metabolismo , Células HEK293 , HumanosRESUMEN
It is now well appreciated that the apoptosome, which governs caspase-dependent cell death, also drives nonapoptotic caspase activation to remodel cells. However, the determinants that specify whether the apoptosome acts to kill or remodel have yet to be identified. Here we report that Tango7 collaborates with the Drosophila apoptosome to drive a caspase-dependent remodeling process needed to resolve individual sperm from a syncytium. In these cells, Tango7 is required for caspase activity and localizes to the active apoptosome compartment via its C terminus. Furthermore, Tango7 directly stimulates the activity of this complex in vitro. We propose that Tango7 specifies the Drosophila apoptosome as an effector of cellular remodeling.
Asunto(s)
Apoptosomas/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Caspasas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Proteínas de Drosophila/genética , Fertilidad/genética , Variación Genética , Masculino , Mutación , Espermatogénesis/genética , Espermatozoides/enzimología , Espermatozoides/metabolismoRESUMEN
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.
Asunto(s)
Citocromos c/metabolismo , Neuroglobina/metabolismo , Animales , Apoptosis , Apoptosomas/metabolismo , Sitios de Unión , Citocromos c/química , Transporte de Electrón , Caballos , Simulación del Acoplamiento Molecular , Neuroglobina/química , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Electricidad EstáticaRESUMEN
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.
Asunto(s)
Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 9/metabolismo , Holoenzimas/metabolismo , Apoptosis , Apoptosomas/química , Apoptosomas/ultraestructura , Factor Apoptótico 1 Activador de Proteasas/química , Factor Apoptótico 1 Activador de Proteasas/genética , Caspasa 9/química , Caspasa 9/genética , Dominio de Reclutamiento y Activación de Caspasas/genética , Microscopía por Crioelectrón , Activación Enzimática , Holoenzimas/química , Holoenzimas/ultraestructura , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Dominios Proteicos , Multimerización de ProteínaRESUMEN
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.
Asunto(s)
Apoptosomas/metabolismo , Técnicas Biosensibles/métodos , Apoptosis/efectos de los fármacos , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Etopósido/farmacología , Células HEK293 , Humanos , Luciferasas/metabolismoRESUMEN
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.
Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Cistatina B/genética , Regulación Neoplásica de la Expresión Génica , Lisosomas/efectos de los fármacos , Vitamina K 3/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/genética , Apoptosomas/efectos de los fármacos , Apoptosomas/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsina B/metabolismo , Catepsina C/antagonistas & inhibidores , Catepsina C/genética , Catepsina C/metabolismo , Catepsina D/antagonistas & inhibidores , Catepsina D/genética , Catepsina D/metabolismo , Catepsinas/antagonistas & inhibidores , Catepsinas/genética , Catepsinas/metabolismo , Cistatina B/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Lisosomas/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Pepstatinas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteasas/farmacología , Proteolisis/efectos de los fármacos , Transducción de Señal , Células U937RESUMEN
Porphyromonas gingivalis is able to invade and modulate host-immune response to promote its survival. This bacterium modulates the cell cycle and programed cell death, contributing to periodontal lesion worsening. Several molecular pathways have been identified as key triggers of apoptosis, including apoptosome apoptotic peptidase activating factor 1 (APAF-1). Apaf-1 and X-linked inhibitor of apoptosis protein (Xiap) mRNA were differentially expressed between gingival samples harvested from human healthy and chronic periodontitis tissues (Apaf-1, 19.2-fold; caspase-9, 14.5-fold; caspase-3, 6.8-fold; Xiap: 2.5-fold in chronic periodontitis) (P < 0.05), highlighting their potential role in periodontitis. An increased proteic expression of APAF-1 was also observed in a murine experimental periodontitis model induced by P. gingivalis-soaked ligatures. In vitro, it was observed that P. gingivalis targets APAF-1, XIAP, caspase-3, and caspase-9, to inhibit epithelial cell death at both mRNA and protein levels. Opposite effect was observed in fibroblasts in which P. gingivalis increased cell death and apoptosis. To assess if the observed effects were associated to APAF-1, epithelial cells and fibroblasts were transfected with siRNA targeting Apaf-1. Herein, we confirmed that APAF-1 is targeted by P. gingivalis in both cell types. This study identified APAF-1 apoptosome and XIAP as intracellular targets of P. gingivalis, contributing to the deterioration of periodontal lesion through an increased persistence of the bacteria within tissues and the subversion of host-immune response.
Asunto(s)
Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/biosíntesis , Infecciones por Bacteroidaceae/metabolismo , Periodontitis Crónica/microbiología , Porphyromonas gingivalis/patogenicidad , Anciano , Animales , Factor Apoptótico 1 Activador de Proteasas/genética , Infecciones por Bacteroidaceae/patología , Células Cultivadas , Periodontitis Crónica/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/fisiología , Encía/metabolismo , Encía/microbiología , Encía/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , ARN Mensajero/genéticaRESUMEN
Apoptosis is an important antiviral host defense mechanism. Here we report the identification of a novel apoptosis inhibitor encoded by the vaccinia virus (VACV) M1L gene. M1L is absent in the attenuated modified vaccinia virus Ankara (MVA) strain of VACV, a strain that stimulates apoptosis in several types of immune cells. M1 expression increased the viability of MVA-infected THP-1 and Jurkat cells and reduced several biochemical hallmarks of apoptosis, such as PARP-1 and procaspase-3 cleavage. Furthermore, ectopic M1L expression decreased staurosporine-induced (intrinsic) apoptosis in HeLa cells. We then identified the molecular basis for M1 inhibitory function. M1 allowed mitochondrial depolarization but blocked procaspase-9 processing, suggesting that M1 targeted the apoptosome. In support of this model, we found that M1 promoted survival in Saccharomyces cerevisiae overexpressing human Apaf-1 and procaspase-9, critical components of the apoptosome, or overexpressing only conformationally active caspase-9. In mammalian cells, M1 coimmunoprecipitated with Apaf-1-procaspase-9 complexes. The current model is that M1 associates with and allows the formation of the apoptosome but prevents apoptotic functions of the apoptosome. The M1 protein features 14 predicted ankyrin (ANK) repeat domains, and M1 is the first ANK-containing protein reported to use this inhibitory strategy. Since ANK-containing proteins are encoded by many large DNA viruses and found in all domains of life, studies of M1 may lead to a better understanding of the roles of ANK proteins in virus-host interactions.IMPORTANCE Apoptosis selectively eliminates dangerous cells such as virus-infected cells. Poxviruses express apoptosis antagonists to neutralize this antiviral host defense. The vaccinia virus (VACV) M1 ankyrin (ANK) protein, a protein with no previously ascribed function, inhibits apoptosis. M1 interacts with the apoptosome and prevents procaspase-9 processing as well as downstream procaspase-3 cleavage in several cell types and under multiple conditions. M1 is the first poxviral protein reported to associate with and prevent the function of the apoptosome, giving a more detailed picture of the threats VACV encounters during infection. Dysregulation of apoptosis is associated with several human diseases. One potential treatment of apoptosis-related diseases is through the use of designed ANK repeat proteins (DARPins), similar to M1, as caspase inhibitors. Thus, the study of the novel antiapoptosis effects of M1 via apoptosome association will be helpful for understanding how to control apoptosis using either natural or synthetic molecules.
Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Apoptosomas/metabolismo , Virus Vaccinia/genética , Animales , Repetición de Anquirina , Apoptosis/efectos de los fármacos , Factor Apoptótico 1 Activador de Proteasas/genética , Caspasa 9/genética , Caspasa 9/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Saccharomyces cerevisiae/genética , Estaurosporina/farmacología , Virus Vaccinia/efectos de los fármacos , Virus Vaccinia/metabolismoRESUMEN
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.
Asunto(s)
Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Apoptosis , Factor Apoptótico 1 Activador de Proteasas/química , Biopolímeros/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Sistema Libre de Células , Cromatografía de Afinidad , Cromatografía en Gel , Activación Enzimática , Células HEK293 , Humanos , Luciferasas/metabolismo , Unión Proteica , Proteolisis , Repeticiones WD40RESUMEN
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.
Asunto(s)
Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Caspasa 9/metabolismo , Apoptosomas/química , Factor Apoptótico 1 Activador de Proteasas/química , Citocromos c/metabolismo , Humanos , Cinética , Modelos MolecularesRESUMEN
Autocatalytic activation of an initiator caspase triggers the onset of apoptosis. In dying cells, caspase-9 activation is mediated by a multimeric adaptor complex known as the Apaf-1 apoptosome. The molecular mechanism by which caspase-9 is activated by the Apaf-1 apoptosome remains largely unknown. Here we demonstrate that the previously reported 1:1 interaction between Apaf-1 caspase recruitment domain (CARD) and caspase-9 CARD is insufficient for the activation of caspase-9. Rather, formation of a multimeric CARD:CARD assembly between Apaf-1 and caspase-9, which requires three types of distinct interfaces, underlies caspase-9 activation. Importantly, an additional surface area on the multimeric CARD assembly is essential for caspase-9 activation. Together, these findings reveal mechanistic insights into the activation of caspase-9 by the Apaf-1 apoptosome and support the induced conformation model for initiator caspase activation by adaptor complexes.
Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Apoptosomas/metabolismo , Caspasa 9/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 9/metabolismo , Catálisis , Activación Enzimática , Humanos , Enlace de Hidrógeno , Técnicas In Vitro , Lisina/química , Modelos Químicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismoRESUMEN
During apoptosis, cytochrome c is released from mitochondria to the cytosol, where it binds Apaf-1. The Apaf-1/cytochrome c complex then oligomerizes either into heptameric caspase-9-activating apoptosome, which subsequently activates caspase-3 and caspase-7, or bigger inactive aggregates, depending on the availability of nucleotide dATP/ATP. A tumor suppressor protein, PHAPI, enhances caspase-9 activation by promoting apoptosome formation through an unknown mechanism. We report here the identification of cellular apoptosis susceptibility protein (CAS) and heat shock protein 70 (Hsp70) as mediators of PHAPI activity. PHAPI, CAS, and Hsp70 function together to accelerate nucleotide exchange on Apaf-1 and prevent inactive Apaf-1/cytochrome c aggregation. CAS expression is induced by multiple apoptotic stimuli including UV irradiation. Knockdown of CAS by RNA interference (RNAi) in cells attenuates apoptosis induced by UV light and causes endogenous Apaf-1 to form aggregates. These studies indicated that PHAPI, CAS, and Hsp70 play an important regulatory role during apoptosis.
Asunto(s)
Apoptosis , Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Proteína de Susceptibilidad a Apoptosis Celular/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor Apoptótico 1 Activador de Proteasas/antagonistas & inhibidores , Caspasa 9/metabolismo , Inhibidores de Caspasas , Proteína de Susceptibilidad a Apoptosis Celular/antagonistas & inhibidores , Proteína de Susceptibilidad a Apoptosis Celular/genética , Activación Enzimática , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares , Nucleótidos/metabolismo , Interferencia de ARN , Proteínas de Unión al ARNRESUMEN
Programmed cell death plays a central role in the regulation of homeostasis and development of multicellular organisms. Deregulation of programmed cell death is connected to a number of disorders, including cancer and autoimmune diseases. Initiation of cell death occurs in the multiprotein complexes or high molecular weight platforms. Composition, structure, and molecular interactions within these platforms influence the cellular decision toward life or death and, therefore, define the induction of a particular cell death program. Here, we discuss in detail the key cell-death complexes-including DISC, complex II, and TNFRI complex I/II, and the necrosome, RIPoptosome, apoptosome, and PIDDosome-that control apoptosis or necroptosis pathways as well as their regulation. The possibility of their pharmacological targeting leading to the development of new strategies of interference with cell death programs via control of the high molecular weight platforms will be discussed.
Asunto(s)
Muerte Celular/fisiología , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Necrosis/fisiopatología , Transducción de Señal/fisiología , Apoptosomas/metabolismo , Caspasa 2/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismoRESUMEN
Differentiation is an inseparable process of development in multicellular organisms. Mouse embryonic stem cells (mESCs) represent a valuable research tool to conduct in vitro studies of cell differentiation. Apoptosis as a well known cell death mechanism shows some common features with cell differentiation, which has caused a number of ambiguities in the field. The research question here is how cells could differentiate these two processes from each other. We have investigated the role of the mitochondrial apoptotic pathway and cell energy level during differentiation of mESCs into the cardiomyocytes and their apoptosis. p53 expression, cytochrome c release, apoptosome formation, and caspase-3/7 activation are observed upon induction of both apoptosis and differentiation. However, remarkable differences are detected in time of cytochrome c appearance, apoptosome formation, and caspase activity upon induction of both processes. In apoptosis, apoptosome formation and caspase activity were observed rapidly following the cytochrome c release. Unlike apoptosis, the release of cytochrome c upon differentiation took more time, and the maximum caspase activity was also postponed for 24 h. This delay suggests that there is a regulatory mechanism during differentiation of mESCs into cardiomyocytes. The highest ATP content of cells was observed immediately after cytochrome c release 6 h after apoptosis induction and then decreased, but it was gradually increased up to 48 h after differentiation. These observations suggest that a delay in the release of cytochrome c or delay in ATP increase attenuate apoptosome formation, and caspase activation thereby discriminates apoptosis from differentiation in mESCs.
Asunto(s)
Apoptosis , Apoptosomas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Caspasas/genética , Caspasas/metabolismo , Línea Celular , Citocromos c/genética , Citocromos c/metabolismo , Células Madre Embrionarias/citología , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Factores de TiempoRESUMEN
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.
Asunto(s)
Apoptosis/genética , Apoptosomas/metabolismo , Factor Apoptótico 1 Activador de Proteasas/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosomas/efectos de los fármacos , Apoptosomas/genética , Factor Apoptótico 1 Activador de Proteasas/genética , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Citocromos c/genética , Citocromos c/metabolismo , Nucleótidos de Desoxiadenina/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Norepinefrina/farmacología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Transducción de Señal , TransfecciónRESUMEN
Apoptosis is a conserved form of programmed cell death firmly established in the aetiology, pathogenesis and treatment of many human diseases. Central to the core machinery of apoptosis are the caspases and their proximal regulators. Current models for caspase control involve a balance of opposing elements, with variable contributions from positive and negative regulators among different cell types and species. To advance a comprehensive view of components that support caspase-dependent cell death, we conducted a genome-wide silencing screen in the Drosophila model. Our strategy used a library of double-stranded RNAs together with a chemical antagonist of Inhibitor of apoptosis proteins (IAPs) that simulates the action of native regulators in the Reaper and Smac (also known as Diablo) families. Here we present a highly validated set of targets that is necessary for death provoked by several stimuli. Among these, Tango7 is identified as a new effector. Cells depleted for this gene resisted apoptosis at a step before the induction of effector caspase activity, and the directed silencing of Tango7 in Drosophila prevented caspase-dependent programmed cell death. Unlike known apoptosis regulators in this model system, Tango7 activity did not influence stimulus-dependent loss of Drosophila DIAP1 (also known as th and IAP1), but instead regulated levels of the apical caspase Dronc (Nc). Similarly, the human Tango7 counterpart, PCID1 (also known as EIF3M), impinged on caspase 9, revealing a new regulatory axis affecting the apoptosome.
Asunto(s)
Apoptosis/genética , Apoptosis/fisiología , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Factores Eucarióticos de Iniciación/metabolismo , Silenciador del Gen , Genoma de los Insectos/genética , Animales , Apoptosomas/metabolismo , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Caspasa 9/metabolismo , Caspasas/metabolismo , Secuencia Conservada , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Factor 3 de Iniciación Eucariótica , Genes de Insecto/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Proteínas Mitocondriales , Imitación Molecular , Interferencia de ARN , ARN Bicatenario/genética , Reproducibilidad de los Resultados , Proteínas de XenopusRESUMEN
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.