Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Intern Med ; 281(5): 458-470, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28425584

RESUMEN

BACKGROUND: Autophagy is a catabolic process involving the engulfment of cytoplasmic content within autophagosomes followed by their delivery to lysosomes. This process is a survival mechanism, enabling cells to cope with nutrient deprivation by degradation and recycling of macromolecules. Yet during continued stress such as prolonged starvation, a switch from autophagy to apoptosis is often detected. OBJECTIVE: In this work, we characterized the temporal dynamics of the transition from autophagy towards apoptosis with the aim of elucidating the molecular mechanism regulating the switch from survival autophagy to apoptotic cell death. RESULTS AND CONCLUSIONS: We defined an inverse relationship between apoptosis and autophagy spanning a period of 72 h, manifested by the sequential reduction in LC3 lipidation and the activation of caspase-3. The transition to apoptosis correlated with a selective decline in the mRNA and protein levels of two anti-apoptotic IAP family proteins, survivin and cIAP2 and a selective increase in the BH3-only protein, BimEL. This 'molecular signature' was common to several cell lines undergoing the switch from autophagy to apoptosis during prolonged starvation. Mechanistically, the increased BimEL protein levels resulted from its reduced binding to its specific E3 ligase, ßTrCP, leading to protein stabilization. Consistent with this, BimEL showed decreased phosphorylation at critical sites previously reported to be essential for binding to the E3 ligase. The decrease in the anti-apoptotic IAPs and the increase in the pro-apoptotic BimEL may thus constitute a molecular switch from autophagy to apoptosis during prolonged starvation.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Proteína 11 Similar a Bcl2/metabolismo , Proteínas Inhibidoras de la Apoptosis/metabolismo , Inanición/fisiopatología , Ubiquitina-Proteína Ligasas/metabolismo , Células A549 , Proteínas Relacionadas con la Autofagia/metabolismo , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Células Cultivadas , Humanos , Survivin , Proteínas con Repetición de beta-Transducina/metabolismo
2.
Cell Death Differ ; 22(3): 465-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25361081

RESUMEN

Autophagy is a tightly regulated catabolic process, which is upregulated in cells in response to many different stress signals. Inhibition of mammalian target of rapmaycin complex 1 (mTORC1) is a crucial step in induction of autophagy, yet the mechanisms regulating the fine tuning of its activity are not fully understood. Here we show that death-associated protein kinase 2 (DAPK2), a Ca(2+)-regulated serine/threonine kinase, directly interacts with and phosphorylates mTORC1, and has a part in suppressing mTOR activity to promote autophagy induction. DAPK2 knockdown reduced autophagy triggered either by amino acid deprivation or by increases in intracellular Ca(2+) levels. At the molecular level, DAPK2 depletion interfered with mTORC1 inhibition caused by these two stresses, as reflected by the phosphorylation status of mTORC1 substrates, ULK1 (unc-51-like kinase 1), p70 ribosomal S6 kinase and eukaryotic initiation factor 4E-binding protein 1. An increase in mTORC1 kinase activity was also apparent in unstressed cells that were depleted of DAPK2. Immunoprecipitated mTORC1 from DAPK2-depleted cells showed increased kinase activity in vitro, an indication that DAPK2 regulation of mTORC1 is inherent to the complex itself. Indeed, we found that DAPK2 associates with components of mTORC1, as demonstrated by co-immunoprecipitation with mTOR and its complex partners, raptor (regulatory-associated protein of mTOR) and ULK1. DAPK2 was also able to interact directly with raptor, as shown by recombinant protein-binding assay. Finally, DAPK2 was shown to phosphorylate raptor in vitro. This phosphorylation was mapped to Ser721, a site located within a highly phosphorylated region of raptor that has previously been shown to regulate mTORC1 activity. Thus, DAPK2 is a novel kinase of mTORC1 and is a potential new member of this multiprotein complex, modulating mTORC1 activity and autophagy levels under stress and steady-state conditions.


Asunto(s)
Autofagia/fisiología , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Fosforilación , Transfección
3.
Cell Death Dis ; 5: e1237, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24853415

RESUMEN

The presence of tangles composed of phosphorylated tau is one of the neuropathological hallmarks of Alzheimer's disease (AD). Tau, a microtubule (MT)-associated protein, accumulates in AD potentially as a result of posttranslational modifications, such as hyperphosphorylation and conformational changes. However, it has not been fully understood how tau accumulation and phosphorylation are deregulated. In the present study, we identified a novel role of death-associated protein kinase 1 (DAPK1) in the regulation of the tau protein. We found that hippocampal DAPK1 expression is markedly increased in the brains of AD patients compared with age-matched normal subjects. DAPK1 overexpression increased tau protein stability and phosphorylation at multiple AD-related sites. In contrast, inhibition of DAPK1 by overexpression of a DAPK1 kinase-deficient mutant or by genetic knockout significantly decreased tau protein stability and abolished its phosphorylation in cell cultures and in mice. Mechanistically, DAPK1-enhanced tau protein stability was mediated by Ser71 phosphorylation of Pin1, a prolyl isomerase known to regulate tau protein stability, phosphorylation, and tau-related pathologies. In addition, inhibition of DAPK1 kinase activity significantly increased the assembly of MTs and accelerated nerve growth factor-mediated neurite outgrowth. Given that DAPK1 has been genetically linked to late onset AD, these results suggest that DAPK1 is a novel regulator of tau protein abundance, and that DAPK1 upregulation might contribute to tau-related pathologies in AD. Therefore, we offer that DAPK1 might be a novel therapeutic target for treating human AD and other tau-related pathologies.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Encéfalo/enzimología , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Neuronas/enzimología , Proteínas tau/metabolismo , Factores de Edad , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Estudios de Casos y Controles , Proteínas Quinasas Asociadas a Muerte Celular/deficiencia , Proteínas Quinasas Asociadas a Muerte Celular/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Mutación , Células 3T3 NIH , Peptidilprolil Isomerasa de Interacción con NIMA , Neuritas/enzimología , Neuritas/patología , Neuronas/patología , Células PC12 , Isomerasa de Peptidilprolil/deficiencia , Isomerasa de Peptidilprolil/genética , Fosforilación , Estabilidad Proteica , Interferencia de ARN , Ratas , Factores de Tiempo , Transfección , Proteínas tau/genética
4.
Oncogene ; 33(5): 611-8, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23318444

RESUMEN

Translational regulation of the p53 mRNA can determine the ratio between p53 and its N-terminal truncated isoforms and therefore has a significant role in determining p53-regulated signaling pathways. Although its importance in cell fate decisions has been demonstrated repeatedly, little is known about the regulatory mechanisms that determine this ratio. Two internal ribosome entry sites (IRESs) residing within the 5'UTR and the coding sequence of p53 mRNA drive the translation of full-length p53 and Δ40p53 isoform, respectively. Here, we report that DAP5, a translation initiation factor shown to positively regulate the translation of various IRES containing mRNAs, promotes IRES-driven translation of p53 mRNA. Upon DAP5 depletion, p53 and Δ40p53 protein levels were decreased, with a greater effect on the N-terminal truncated isoform. Functional analysis using bicistronic vectors driving the expression of a reporter gene from each of these two IRESs indicated that DAP5 preferentially promotes translation from the second IRES residing in the coding sequence. Furthermore, p53 mRNA expressed from a plasmid carrying this second IRES was selectively shifted to lighter polysomes upon DAP5 knockdown. Consequently, Δ40p53 protein levels and the subsequent transcriptional activation of the 14-3-3σ gene, a known target of Δ40p53, were strongly reduced. In addition, we show here that DAP5 interacts with p53 IRES elements in in vitro and in vivo binding studies, proving for the first time that DAP5 directly binds a target mRNA. Thus, through its ability to regulate IRES-dependent translation of the p53 mRNA, DAP5 may control the ratio between different p53 isoforms encoded by a single mRNA.


Asunto(s)
Factor 4G Eucariótico de Iniciación/metabolismo , Iniciación de la Cadena Peptídica Traduccional/genética , Isoformas de Proteínas/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteínas 14-3-3/biosíntesis , Proteínas 14-3-3/metabolismo , Regiones no Traducidas 5'/genética , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Factor 4G Eucariótico de Iniciación/genética , Exorribonucleasas/biosíntesis , Exorribonucleasas/metabolismo , Regulación de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Isoformas de Proteínas/biosíntesis , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Ribosomas/metabolismo , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética
5.
Oncogene ; 31(6): 683-93, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21725354

RESUMEN

Death-associated protein kinase (DAPk), a multi-domain serine/threonine kinase, regulates numerous cell death mechanisms and harbors tumor suppressor functions. In this study, we report that DAPk directly binds and functionally activates pyruvate kinase M2 (PKM2), a key glycolytic enzyme, which contributes to the regulation of cancer cell metabolism. PKM2 was identified as a novel binding partner of DAPk by a yeast two-hybrid screen. This interaction was validated in vitro by enzyme-linked immunosorbent assay using purified proteins and in vivo by co-immunoprecipitation of the two endogenous proteins from cells. In vitro interaction with full-length DAPk resulted in a significant increase in the activity of PKM2. Conversely, a fragment of DAPk harboring only the functional kinase domain (KD) could neither bind PKM2 in cells nor activate it in vitro. Indeed, DAPk failed to phosphorylate PKM2. Notably, transfection of cells, with a truncated DAPk lacking the KD, elevated endogenous PKM2 activity, suggesting that PKM2 activation by DAPk occurs independently of its kinase activity. DAPk-transfected cells displayed changes in glycolytic activity, as reflected by elevated lactate production, whereas glucose uptake remained unaltered. A mild reduction in cell proliferation was detected as well in these transfected cells. Altogether, this work identifies a new role for DAPk as a metabolic regulator, suggesting the concept of direct interactions between a tumor suppressor and a key glycolytic enzyme to limit cell growth. Moreover, the work documents a unique function of DAPk that is independent of its catalytic activity and a novel mechanism to activate PKM2 by protein-protein interaction.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Glucólisis , Piruvato Quinasa/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Sitios de Unión/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular , Activación Enzimática , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Mutación , Fosforilación , Unión Proteica , Piruvato Quinasa/genética , Interferencia de ARN , Técnicas del Sistema de Dos Híbridos
6.
Cell Death Differ ; 19(5): 788-97, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22095288

RESUMEN

Autophagy, a process in which cellular components are engulfed and degraded within double-membrane vesicles termed autophagosomes, has an important role in the response to oxidative damage. Here we identify a novel cascade of phosphorylation events, involving a network of protein and lipid kinases, as crucial components of the signaling pathways that regulate the induction of autophagy under oxidative stress. Our findings show that both the tumor-suppressor death-associated protein kinase (DAPk) and protein kinase D (PKD), which we previously showed to be phosphorylated and consequently activated by DAPk, mediate the induction of autophagy in response to oxidative damage. Furthermore, we map the position of PKD within the autophagic network to Vps34, a lipid kinase whose function is indispensable for autophagy, and demonstrate that PKD is found in the same molecular complex with Vps34. PKD phosphorylates Vps34, leading to activation of Vps34, phosphatydilinositol-3-phosphate (PI(3)P) formation, and autophagosome formation. Consistent with its identification as a novel inducer of the autophagic machinery, we show that PKD is recruited to LC3-positive autophagosomes, where it localizes specifically to the autophagosomal membranes. Taken together, our results describe PKD as a novel Vps34 kinase that functions as an effecter of autophagy under oxidative stress.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Línea Celular , Fosfatidilinositol 3-Quinasas Clase III/genética , Proteínas Quinasas Asociadas a Muerte Celular , Electroforesis en Gel de Poliacrilamida , Inmunoprecipitación , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Proteína Quinasa C/genética , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo
7.
Cell Death Differ ; 18(9): 1507-20, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21311567

RESUMEN

Death-associated protein kinase (DAPK) is a key player in several modes of neuronal death/injury and has been implicated in the late-onset Alzheimer's disease (AD). DAPK promotes cell death partly through its effect on regulating actin cytoskeletons. In this study, we report that DAPK inhibits microtubule (MT) assembly by activating MARK/PAR-1 family kinases MARK1/2, which destabilize MT by phosphorylating tau and related MAP2/4. DAPK death domain, but not catalytic activity, is responsible for this activation by binding to MARK1/2 spacer region, thereby disrupting an intramolecular interaction that inhibits MARK1/2. Accordingly, DAPK(-/-) mice brain displays a reduction of tau phosphorylation and DAPK enhances the effect of MARK2 on regulating polarized neurite outgrowth. Using a well-characterized Drosophila model of tauopathy, we show that DAPK exerts an effect in part through MARK Drosophila ortholog PAR-1 to induce rough eye and loss of photoreceptor neurons. Furthermore, DAPK enhances tau toxicity through a PAR-1 phosphorylation-dependent mechanism. Together, our study reveals a novel mechanism of MARK activation, uncovers DAPK functions in modulating MT assembly and neuronal differentiation, and provides a molecular link of DAPK to tau phosphorylation, an event associated with AD pathology.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Sistema de Señalización de MAP Quinasas , Microtúbulos/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Animales , Diferenciación Celular , Línea Celular , Proteínas Quinasas Asociadas a Muerte Celular , Drosophila , Activación Enzimática , Humanos , Ratones , Microtúbulos/ultraestructura , Neuronas/ultraestructura , Fosforilación
8.
Cell Death Differ ; 17(8): 1244-53, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20150916

RESUMEN

The mammalian cell death network comprises three distinct functional modules: apoptosis, autophagy and programmed necrosis. Currently, the field lacks systems level approaches to assess the extent to which the intermodular connectivity affects cell death performance. Here, we developed a platform that is based on single and double sets of RNAi-mediated perturbations targeting combinations of apoptotic and autophagic genes. The outcome of perturbations is measured both at the level of the overall cell death responses, using an unbiased quantitative reporter, and by assessing the molecular responses within the different functional modules. Epistatic analyses determine whether seemingly unrelated pairs of proteins are genetically linked. The initial running of this platform in etoposide-treated cells, using a few single and double perturbations, identified several levels of connectivity between apoptosis and autophagy. The knock down of caspase3 turned on a switch toward autophagic cell death, which requires Atg5 or Beclin-1. In addition, a reciprocal connection between these two autophagic genes and apoptosis was identified. By applying computational tools that are based on mining the protein-protein interaction database, a novel biochemical pathway connecting between Atg5 and caspase3 is suggested. Scaling up this platform into hundreds of perturbations potentially has a wide, general scope of applicability, and will provide the basis for future modeling of the cell death network.


Asunto(s)
Apoptosis , Autofagia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Bases de Datos de Proteínas , Etopósido/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
9.
Cell Death Differ ; 16(8): 1093-107, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19373242

RESUMEN

Cell death is essential for a plethora of physiological processes, and its deregulation characterizes numerous human diseases. Thus, the in-depth investigation of cell death and its mechanisms constitutes a formidable challenge for fundamental and applied biomedical research, and has tremendous implications for the development of novel therapeutic strategies. It is, therefore, of utmost importance to standardize the experimental procedures that identify dying and dead cells in cell cultures and/or in tissues, from model organisms and/or humans, in healthy and/or pathological scenarios. Thus far, dozens of methods have been proposed to quantify cell death-related parameters. However, no guidelines exist regarding their use and interpretation, and nobody has thoroughly annotated the experimental settings for which each of these techniques is most appropriate. Here, we provide a nonexhaustive comparison of methods to detect cell death with apoptotic or nonapoptotic morphologies, their advantages and pitfalls. These guidelines are intended for investigators who study cell death, as well as for reviewers who need to constructively critique scientific reports that deal with cellular demise. Given the difficulties in determining the exact number of cells that have passed the point-of-no-return of the signaling cascades leading to cell death, we emphasize the importance of performing multiple, methodologically unrelated assays to quantify dying and dead cells.


Asunto(s)
Muerte Celular , Apoptosis , Células Eucariotas/citología , Citometría de Flujo , Guías como Asunto , Humanos , Immunoblotting , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Espectrometría de Fluorescencia
10.
Cell Death Differ ; 16(7): 966-75, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19325568

RESUMEN

It is not surprising that the demise of a cell is a complex well-controlled process. Apoptosis, the first genetically programmed death process identified, has been extensively studied and its contribution to the pathogenesis of disease well documented. Yet, apoptosis does not function alone to determine a cell's fate. More recently, autophagy, a process in which de novo-formed membrane-enclosed vesicles engulf and consume cellular components, has been shown to engage in a complex interplay with apoptosis. In some cellular settings, it can serve as a cell survival pathway, suppressing apoptosis, and in others, it can lead to death itself, either in collaboration with apoptosis or as a back-up mechanism when the former is defective. The molecular regulators of both pathways are inter-connected; numerous death stimuli are capable of activating either pathway, and both pathways share several genes that are critical for their respective execution. The cross-talk between apoptosis and autophagy is therefore quite complex, and sometimes contradictory, but surely critical to the overall fate of the cell. Furthermore, the cross-talk is a key factor in the outcome of death-related pathologies such as cancer, its development and treatment.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Neoplasias/metabolismo , Transducción de Señal/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Factor de Transcripción E2F1/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Transporte Vesicular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA