Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
Oncogene ; 43(15): 1098-1112, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38388710

RESUMEN

The non-canonical translation initiation factor EIF4G2 plays essential roles in cellular stress responses via translation of selective mRNA cohorts. Currently there is limited and conflicting information regarding its involvement in cancer development and progression. Here we assessed its role in endometrial cancer (EC), in a cohort of 280 EC patients across different types, grades, and stages, and found that low EIF4G2 expression highly correlated with poor overall- and recurrence-free survival in Grade 2 EC patients, monitored over a period of up to 12 years. To establish a causative connection between low EIF4G2 expression and cancer progression, we stably knocked-down EIF4G2 in two human EC cell lines in parallel. EIF4G2 depletion resulted in increased resistance to conventional therapies and increased the prevalence of molecular markers for aggressive cell subsets, altering their transcriptional and proteomic landscapes. Prominent among the proteins with decreased abundance were Kinesin-1 motor proteins, KIF5B and KLC1, 2, 3. Multiplexed imaging of the EC patient tumor cohort showed a correlation between decreased expression of the kinesin proteins, and poor survival in patients with tumors of certain grades and stages. These findings reveal potential novel biomarkers for Grade 2 EC with ramifications for patient stratification and therapeutic interventions.


Asunto(s)
Neoplasias Endometriales , Cinesinas , Femenino , Humanos , Cinesinas/genética , Proteómica , Línea Celular , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Factor 4G Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/metabolismo
2.
Life Sci Alliance ; 7(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38129098

RESUMEN

Tumor cells often exploit the protein translation machinery, resulting in enhanced protein expression essential for tumor growth. Since canonical translation initiation is often suppressed because of cell stress in the tumor microenvironment, non-canonical translation initiation mechanisms become particularly important for shaping the tumor proteome. EIF4G2 is a non-canonical translation initiation factor that mediates internal ribosome entry site (IRES)- and uORF-dependent initiation mechanisms, which can be used to modulate protein expression in cancer. Here, we explored the contribution of EIF4G2 to cancer by screening the COSMIC database for EIF4G2 somatic mutations in cancer patients. Functional examination of missense mutations revealed deleterious effects on EIF4G2 protein-protein interactions and, importantly, on its ability to mediate non-canonical translation initiation. Specifically, one mutation, R178Q, led to reductions in protein expression and near-complete loss of function. Two other mutations within the MIF4G domain specifically affected EIF4G2's ability to mediate IRES-dependent translation initiation but not that of target mRNAs with uORFs. These results shed light on both the structure-function of EIF4G2 and its potential tumor suppressor effects.


Asunto(s)
Neoplasias , Biosíntesis de Proteínas , Humanos , Biosíntesis de Proteínas/genética , Mutación/genética , Neoplasias/genética , Factor 4G Eucariótico de Iniciación/genética , Microambiente Tumoral
3.
Autophagy ; 19(8): 2372-2385, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37184247

RESUMEN

Macroautophagy/autophagy is a catabolic process by which cytosolic content is engulfed, degraded and recycled. It has been implicated as a critical pathway in advanced stages of cancer, as it maintains tumor cell homeostasis and continuous growth by nourishing hypoxic or nutrient-starved tumors. Autophagy also supports alternative cellular trafficking pathways, providing a mechanism of non-canonical secretion of inflammatory cytokines. This opens a significant therapeutic opportunity for using autophagy inhibitors in cancer and acute inflammatory responses. Here we developed a high throughput compound screen to identify inhibitors of protein-protein interaction (PPI) in autophagy, based on the protein-fragment complementation assay (PCA). We chose to target the ATG12-ATG3 PPI, as this interaction is indispensable for autophagosome formation, and the analyzed structure of the interaction interface predicts that it may be amenable to inhibition by small molecules. We screened 41,161 compounds yielding 17 compounds that effectively inhibit the ATG12-ATG3 interaction in the PCA platform, and which were subsequently filtered by their ability to inhibit autophagosome formation in viable cells. We describe a lead compound (#189) that inhibited GFP-fused MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta) puncta formation in cells with IC50 value corresponding to 9.3 µM. This compound displayed a selective inhibitory effect on the growth of autophagy addicted tumor cells and inhibited secretion of IL1B/IL-1ß (interleukin 1 beta) by macrophage-like cells. Compound 189 has the potential to be developed into a therapeutic drug and its discovery documents the power of targeting PPIs for acquiring specific and selective compound inhibitors of autophagy.Abbreviations: ANOVA: analysis of variance; ATG: autophagy related; CQ: chloroquine; GFP: green fluorescent protein; GLuc: Gaussia Luciferase; HEK: human embryonic kidney; IL1B: interleukin 1 beta; LPS: lipopolysaccharide; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; PCA: protein-fragment complementation assay; PDAC: pancreatic ductal adenocarcinoma; PMA: phorbol 12-myristate 13-acetate; PPI: protein-protein interaction. VCL: vinculin.


Asunto(s)
Autofagia , Neoplasias Pancreáticas , Humanos , Interleucina-1beta/farmacología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Relacionadas con la Autofagia , Proteínas Fluorescentes Verdes/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteína 12 Relacionada con la Autofagia
4.
RNA ; 28(10): 1325-1336, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35961752

RESUMEN

Death associated protein 5 (DAP5/eIF4G2/NAT1) is a member of the eIF4G translation initiation factors that has been shown to mediate noncanonical and/or cap-independent translation. It is essential for embryonic development and for differentiation of embryonic stem cells (ESCs), specifically its ability to drive translation of specific target mRNAs. In order to expand the repertoire of DAP5 target mRNAs, we compared ribosome profiles in control and DAP5 knockdown (KD) human ESCs (hESCs) to identify mRNAs with decreased ribosomal occupancy upon DAP5 silencing. A cohort of 68 genes showed decreased translation efficiency in DAP5 KD cells. Mass spectrometry confirmed decreased protein abundance of a significant portion of these targets. Among these was KMT2D, a histone methylase previously shown to be essential for ESC differentiation and embryonic development. We found that nearly half of the cohort of DAP5 target mRNAs displaying reduced translation efficiency of their main coding sequences upon DAP5 KD contained upstream open reading frames (uORFs) that are actively translated independently of DAP5. This is consistent with previously suggested mechanisms by which DAP5 mediates leaky scanning through uORFs and/or reinitiation at the main coding sequence. Crosslinking protein-RNA immunoprecipitation experiments indicated that a significant subset of DAP5 mRNA targets bound DAP5, indicating that direct binding between DAP5 protein and its target mRNAs is a frequent but not absolute requirement for DAP5-dependent translation of the main coding sequence. Thus, we have extended DAP5's function in translation of specific mRNAs in hESCs by a mechanism allowing translation of the main coding sequence following upstream translation of short ORFs.


Asunto(s)
Factor 4G Eucariótico de Iniciación/metabolismo , Células Madre Embrionarias Humanas , Histona Metiltransferasas/genética , Histona Metiltransferasas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Sistemas de Lectura Abierta/genética , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
5.
Mol Oncol ; 16(6): 1365-1383, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122388

RESUMEN

Radiation therapy can induce cellular senescence in cancer cells, leading to short-term tumor growth arrest but increased long-term recurrence. To better understand the molecular mechanisms involved, we developed a model of radiation-induced senescence in cultured cancer cells. The irradiated cells exhibited a typical senescent phenotype, including upregulation of p53 and its main target, p21, followed by a sustained reduction in cellular proliferation, changes in cell size and cytoskeleton organization, and senescence-associated beta-galactosidase activity. Mass spectrometry-based proteomic profiling of the senescent cells indicated downregulation of proteins involved in cell cycle progression and DNA repair, and upregulation of proteins associated with malignancy. A functional siRNA screen using a cell death-related library identified mitochondrial serine protease HtrA2 as being necessary for sustained growth arrest of the senescent cells. In search of direct HtrA2 substrates following radiation, we determined that HtrA2 cleaves the intermediate filament protein vimentin, affecting its cytoplasmic organization. Ectopic expression of active cytosolic HtrA2 resulted in similar changes to vimentin filament assembly. Thus, HtrA2 is involved in the cytoskeletal reorganization that accompanies radiation-induced senescence and the continuous maintenance of proliferation arrest.


Asunto(s)
Senescencia Celular , Serina Peptidasa A2 que Requiere Temperaturas Altas , Neoplasias , Proteómica , Apoptosis , Senescencia Celular/fisiología , Senescencia Celular/efectos de la radiación , Serina Peptidasa A2 que Requiere Temperaturas Altas/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas/metabolismo , Humanos , Proteínas Mitocondriales/metabolismo , Neoplasias/genética , Neoplasias/radioterapia , Células Tumorales Cultivadas , Vimentina/metabolismo
6.
Cell Death Differ ; 29(6): 1255-1266, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34992231

RESUMEN

The role of programmed cell death during embryonic development has been described previously, but its specific contribution to peri- and post-implantation stages is still debatable. Here, we used transmission electron microscopy and immunostaining of E5.5-7.5 mouse embryos to investigate death processes during these stages of development. We report that in addition to canonical apoptosis observed in E5.5-E7.5 embryos, a novel type of cell elimination occurs in E7.5 embryos among the epiblasts at the apical side, in which cells shed membrane-enclosed fragments of cytosol and organelles into the lumen, leaving behind small, enucleated cell remnants at the apical surface. This process is caspase-independent as it occurred in Apaf1 knockout embryos. We suggest that this novel mechanism controls epiblast cell numbers. Altogether, this work documents the activation of two distinct programs driving irreversible terminal states of epiblast cells in the post-implantation mouse embryo.


Asunto(s)
Desarrollo Embrionario , Estratos Germinativos , Animales , Apoptosis , Implantación del Embrión , Embrión de Mamíferos/metabolismo , Femenino , Ratones , Embarazo
7.
Cell Death Dis ; 11(5): 305, 2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366830

RESUMEN

Death-associated protein kinase 1 (DAPK1, DAPk, DAPK) is known for its involvement in apoptosis and autophagy-associated cell death. Here, we identified an unexpected function of DAPK1 in suppressing necroptosis. DAPK1-deficiency renders macrophages and dendritic cells susceptible to necroptotic death. We also observed an inhibitory role for DAPK1 in necroptosis in HT-29 cells, since knockdown or knockout of DAPK1 in such cells increased their sensitivity to necroptosis. Increased necroptosis was associated with enhanced formation of the RIPK1-RIPK3-MLKL complex in these DAPK1-deficient cells. We further found that DAPK1-deficiency led to decreased MAPK activated kinase 2 (MK2) activation and reduced RIPK1 S321 phosphorylation, with this latter representing a critical step controlling necrosome formation. Most TNF signaling pathways, including ERK, JNK, and AKT, were not regulated by DAPK. In contrast, DAPK bound p38 MAPK and selectively promoted p38 MAPK activation, resulting in enhanced MK2 phosphorylation. Our results reveal a novel role for DAPK1 in inhibiting necroptosis and illustrate an unexpected selectivity for DAPK1 in promoting p38 MAPK-MK2 activation. Importantly, our study suggests that modulation of necroptosis and p38/MK2-mediated inflammation may be achieved by targeting DAPK1.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Necroptosis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Caspasa 8/metabolismo , Supervivencia Celular , Proteínas Quinasas Asociadas a Muerte Celular/deficiencia , Regulación hacia Abajo , Activación Enzimática , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/enzimología , Células Mieloides/patología , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Choque Séptico/metabolismo , Choque Séptico/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa
8.
Cell Rep ; 29(1): 104-117.e4, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31577941

RESUMEN

The glucocorticoid receptor (GR) acts as a ubiquitous cortisol-dependent transcription factor (TF). To identify co-factors, we used protein-fragment complementation assays and found that GR recognizes FLI1 and additional ETS family proteins, TFs relaying proliferation and/or migration signals. Following steroid-dependent translocation of FLI1 and GR to the nucleus, the FLI1-specific domain (FLS) binds with GR and strongly enhances GR's transcriptional activity. This interaction has functional consequences in Ewing sarcoma (ES), childhood and adolescence bone malignancies driven by fusions between EWSR1 and FLI1. In vitro, GR knockdown inhibited the migration and proliferation of ES cells, and in animal models, antagonizing GR (or lowering cortisol) retarded both tumor growth and metastasis from bone to lung. Taken together, our findings offer mechanistic rationale for repurposing GR-targeting drugs for the treatment of patients with ES.


Asunto(s)
Proteínas Proto-Oncogénicas c-ets/metabolismo , Receptores de Glucocorticoides/metabolismo , Sarcoma de Ewing/metabolismo , Animales , Neoplasias Óseas/metabolismo , Movimiento Celular/fisiología , Núcleo Celular/metabolismo , Proliferación Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Células HEK293 , Humanos , Ratones , Ratones SCID , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo
9.
Autophagy ; 15(10): 1829-1833, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31234750

RESUMEN

The NIH-funded center for autophagy research named Autophagy, Inflammation, and Metabolism (AIM) Center of Biomedical Research Excellence, located at the University of New Mexico Health Science Center is now completing its second year as a working center with a mission to promote autophagy research locally, nationally, and internationally. The center has thus far supported a cadre of 6 junior faculty (mentored PIs; mPIs) at a near-R01 level of funding. Two mPIs have graduated by obtaining their independent R01 funding and 3 of the remaining 4 have won significant funding from NIH in the form of R21 and R56 awards. The first year and a half of setting up the center has been punctuated by completion of renovations and acquisition and upgrades for equipment supporting autophagy, inflammation and metabolism studies. The scientific cores usage, and the growth of new studies is promoted through pilot grants and several types of enablement initiatives. The intent to cultivate AIM as a scholarly hub for autophagy and related studies is manifested in its Vibrant Campus Initiative, and the Tuesday AIM Seminar series, as well as by hosting a major scientific event, the 2019 AIM symposium, with nearly one third of the faculty from the International Council of Affiliate Members being present and leading sessions, giving talks, and conducting workshop activities. These and other events are often videostreamed for a worldwide scientific audience, and information about events at AIM and elsewhere are disseminated on Twitter and can be followed on the AIM web site. AIM intends to invigorate research on overlapping areas between autophagy, inflammation and metabolism with a number of new initiatives to promote metabolomic research. With the turnover of mPIs as they obtain their independent funding, new junior faculty are recruited and appointed as mPIs. All these activities are in keeping with AIM's intention to enable the next generation of autophagy researchers and help anchor, disseminate, and convey the depth and excitement of the autophagy field.


Asunto(s)
Autofagia/fisiología , Investigación Biomédica/organización & administración , Inflamación , Metabolismo/fisiología , Sociedades Científicas , Investigación Biomédica/economía , Investigación Biomédica/tendencias , Docentes Médicos/economía , Docentes Médicos/educación , Financiación Gubernamental , Organización de la Financiación/economía , Historia del Siglo XXI , Humanos , Inflamación/etiología , Inflamación/patología , Mentores , National Institutes of Health (U.S.)/economía , New Mexico , Investigadores/economía , Investigadores/educación , Sociedades Científicas/economía , Sociedades Científicas/organización & administración , Sociedades Científicas/normas , Sociedades Científicas/tendencias , Estados Unidos
10.
Cell Cycle ; 18(11): 1169-1176, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31116076

RESUMEN

DAPK1 and DAPK2 are calmodulin (CaM)-regulated protein kinases that share a high degree of homology in their catalytic and CaM regulatory domains. Both kinases function as tumor suppressors, and both have been implicated in autophagy regulation. Over the years, common regulatory mechanisms for the two kinases as well as kinase-specific ones have been identified. In a recent work, we revealed that DAPK2 is phosphorylated on Ser289 by the metabolic sensor AMPK, and that this phosphorylation enhances DAPK2 catalytic activity. Notably, Ser289 is conserved between DAPK1 and DAPK2, and was previously found to be phosphorylated in DAPK1 by RSK. Intriguingly, Ser289 phosphorylation was conversely reported to inhibit the pro-apoptotic activity of DAPK1 in cells. However, as the direct effect of this phosphorylation on DAPK1 catalytic activity was not tested, indirect effects were not excluded. Here, we compared Ser289 phosphorylation of the two kinases in the same cells and found that the intracellular signaling pathways that lead to Ser289 phosphorylation are mutually-exclusive and different for each kinase. In addition, we found that Ser289 phosphorylation in fact enhances DAPK1 catalytic activity, similar to the effect on DAPK2. Thus, Ser289 phosphorylation activates both DAPK1 and DAPK2, but in response to different intracellular signaling pathways.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/metabolismo , Catálisis , Proteínas Quinasas Asociadas a Muerte Celular/química , Activación Enzimática , Células HCT116 , Células HEK293 , Humanos , Fosforilación/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Transducción de Señal/fisiología
11.
Autophagy ; 15(3): 438-452, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30238850

RESUMEN

Macroautophagy/autophagy is a conserved catabolic process that maintains cellular homeostasis under basal growth and stress conditions. In cancer, autophagy can either prevent or promote tumor growth, at early or advanced stages, respectively. We screened public databases to identify autophagy-related somatic mutations in cancer, using a computational approach to identify cancer mutational target sites, employing exact statistics. The top significant hit was a missense mutation (Y113C) in the MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta) protein, which occurred at a significant frequency in cancer, and was detected in early stages in primary tumors of patients with known tumor lineage. The mutation reduced the formation of GFP-LC3B puncta and attenuated LC3B lipidation during Torin1-induced autophagy. Its effect on the direct physical interaction of LC3B with each of the 4 proteins that control its maturation or lipidation was tested by applying a protein-fragment complementation assay and co-immunoprecipitation experiments. Interactions with ATG4A and ATG4B proteases were reduced, yet without perturbing the cleavage of mutant LC3B. Most importantly, the mutation significantly reduced the interaction with the E1-like enzyme ATG7, but not the direct interaction with the E2-like enzyme ATG3, suggesting a selective perturbation in the binding of LC3B to some of its partner proteins. Structure analysis and molecular dynamics simulations of LC3B protein and its mutant suggest that the mutation changes the conformation of a loop that has several contact sites with ATG4B and the ATG7 homodimer. We suggest that this loss-of-function mutation, which attenuates autophagy, may promote early stages of cancer development.


Asunto(s)
Proteína 7 Relacionada con la Autofagia/metabolismo , Autofagia/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neoplasias/genética , Autofagia/efectos de los fármacos , Proteína 7 Relacionada con la Autofagia/química , Proteína 7 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Biología Computacional , Cisteína Endopeptidasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/química , Mutación Missense , Naftiridinas/farmacología , Enzimas Ubiquitina-Conjugadoras/metabolismo
12.
Oncotarget ; 9(60): 31570-31571, 2018 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-30167079
13.
J Cell Sci ; 131(18)2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30237248

RESUMEN

Autophagy as a means of cell killing was first advanced by Clark's phenotypic description of 'Type II autophagic cell death' in 1990. However, this phenomenon later came into question, because the presence of autophagosomes in dying cells does not necessarily signify that autophagy is the cause of demise, but rather may reflect the efforts of the cell to prevent it. Resolution of this issue comes from a more careful definition of autophagy-dependent cell death (ADCD) as a regulated cell death that is shown experimentally to require different components of the autophagy machinery without involvement of alternative cell death pathways. Following these strict criteria, ADCD has been validated in both lower model organisms and mammalian cells, highlighting its importance for developmental and pathophysiological cell death. Recently, researchers have defined additional morphological criteria that characterize ADCD and begun to explore how the established, well-studied autophagy pathway is subverted from a survival to a death function. This Review explores validated models of ADCD and focuses on the current understanding of the mechanisms by which autophagy can kill a cell.


Asunto(s)
Autofagia/fisiología , Muerte Celular/fisiología , Humanos
14.
Autophagy ; 14(6): 925-929, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29938597

RESUMEN

Recently, NIH has funded a center for autophagy research named the Autophagy, Inflammation, and Metabolism (AIM) Center of Biomedical Research Excellence, located at the University of New Mexico Health Science Center (UNM HSC), with aspirations to promote autophagy research locally, nationally, and internationally. The center has 3 major missions: (i) to support junior faculty in their endeavors to develop investigations in this area and obtain independent funding; (ii) to develop and provide technological platforms to advance autophagy research with emphasis on cellular approaches for high quality reproducible research; and (iii) to foster international collaborations through the formation of an International Council of Affiliate Members and through hosting national and international workshops and symposia. Scientifically, the AIM center is focused on autophagy and its intersections with other processes, with emphasis on both fundamental discoveries and applied translational research.


Asunto(s)
Autofagia , Investigación Biomédica , Inflamación/patología , Cooperación Internacional , Investigadores , Congresos como Asunto , Difusión de la Información
15.
Nat Commun ; 9(1): 1759, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717115

RESUMEN

Autophagy is an intracellular degradation process essential for adaptation to metabolic stress. DAPK2 is a calmodulin-regulated protein kinase, which has been implicated in autophagy regulation, though the mechanism is unclear. Here, we show that the central metabolic sensor, AMPK, phosphorylates DAPK2 at a critical site in the protein structure, between the catalytic and the calmodulin-binding domains. This phosphorylation activates DAPK2 by functionally mimicking calmodulin binding and mitigating an inhibitory autophosphorylation, providing a novel, alternative mechanism for DAPK2 activation during metabolic stress. In addition, we show that DAPK2 phosphorylates the core autophagic machinery protein, Beclin-1, leading to dissociation of its inhibitor, Bcl-XL. Importantly, phosphorylation of DAPK2 by AMPK enhances DAPK2's ability to phosphorylate Beclin-1, and depletion of DAPK2 reduces autophagy in response to AMPK activation. Our study reveals a unique calmodulin-independent mechanism for DAPK2 activation, critical to its function as a novel downstream effector of AMPK in autophagy.


Asunto(s)
Adenilato Quinasa/metabolismo , Autofagia , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Estrés Fisiológico , Células A549 , Secuencia de Aminoácidos , Animales , Beclina-1/metabolismo , Catálisis , Proteínas Quinasas Asociadas a Muerte Celular/química , Dimerización , Células HCT116 , Células HEK293 , Humanos , Masculino , Ratones , Mutación , Fosforilación , Homología de Secuencia de Aminoácido , Serina/metabolismo , Treonina/metabolismo , Proteína bcl-X/metabolismo
16.
Cell Cycle ; 16(21): 2003-2010, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28933588

RESUMEN

Autophagy is critical for homeostasis and cell survival during stress, but can also lead to cell death, a little understood process that has been shown to contribute to developmental cell death in lower model organisms, and to human cancer cell death. We recently reported 1 on our thorough molecular and morphologic characterization of an autophagic cell death system involving resveratrol treatment of lung carcinoma cells. To gain mechanistic insight into this death program, we performed a signalome-wide RNAi screen for genes whose functions are necessary for resveratrol-induced death. The screen identified GBA1, the gene encoding the lysosomal enzyme glucocerebrosidase, as an important mediator of autophagic cell death. Here we further show the physiological relevance of GBA1 to developmental cell death in midgut regression during Drosophila metamorphosis. We observed a delay in midgut cell death in two independent Gba1a RNAi lines, indicating the critical importance of Gba1a for midgut development. Interestingly, loss-of-function GBA1 mutations lead to Gaucher Disease and are a significant risk factor for Parkinson Disease, which have been associated with defective autophagy. Thus GBA1 is a conserved element critical for maintaining proper levels of autophagy, with high levels leading to autophagic cell death.


Asunto(s)
Autofagia/fisiología , Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/metabolismo , Lisosomas/metabolismo , Animales , Drosophila/metabolismo , Humanos , Lisosomas/ultraestructura , Enfermedad de Parkinson/genética
17.
Cell Death Differ ; 24(7): 1288-1302, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28574511

RESUMEN

Activating alternative cell death pathways, including autophagic cell death, is a promising direction to overcome the apoptosis resistance observed in various cancers. Yet, whether autophagy acts as a death mechanism by over consumption of intracellular components is still controversial and remains undefined at the ultrastructural and the mechanistic levels. Here we identified conditions under which resveratrol-treated A549 lung cancer cells die by a mechanism that fulfills the previous definition of autophagic cell death. The cells displayed a strong and sustained induction of autophagic flux, cell death was prevented by knocking down autophagic genes and death occurred in the absence of apoptotic or necroptotic pathway activation. Detailed ultrastructural characterization revealed additional critical events, including a continuous increase over time in the number of autophagic vacuoles, in particular autolysosomes, occupying most of the cytoplasm at terminal stages. This was followed by loss of organelles, disruption of intracellular membranes including the swelling of perinuclear space and, occasionally, a unique type of nuclear shedding. A signalome-wide shRNA-based viability screen was applied to identify positive mediators of this type of autophagic cell death. One top hit was GBA1, the Gaucher disease-associated gene, which encodes glucocerebrosidase, an enzyme that metabolizes glucosylceramide to ceramide and glucose. Interestingly, glucocerebrosidase expression levels and activity were elevated, concomitantly with increased intracellular ceramide levels, both of which correlated in time with the appearance of the unique death characteristics. Transfection with siGBA1 attenuated the increase in glucocerebrosidase activity and the intracellular ceramide levels. Most importantly, GBA1 knockdown prevented the strong increase in LC3 lipidation, and many of the ultrastructural changes characteristic of this type of autophagic cell death, including a significant decrease in cytoplasmic area occupied by autophagic vacuoles. Together, these findings highlight the critical role of GBA1 in mediating enhanced self-consumption of intracellular components and endomembranes, leading to autophagic cell death.


Asunto(s)
Autofagia , Glucosilceramidasa/metabolismo , Interferencia de ARN , Transducción de Señal , Células A549 , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Necrosis , Interferencia de ARN/efectos de los fármacos , Resveratrol , Transducción de Señal/efectos de los fármacos , Esfingolípidos/metabolismo , Estilbenos/farmacología
18.
Genes Dev ; 30(17): 1991-2004, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27664238

RESUMEN

Multiple transcriptional and epigenetic changes drive differentiation of embryonic stem cells (ESCs). This study unveils an additional level of gene expression regulation involving noncanonical, cap-independent translation of a select group of mRNAs. This is driven by death-associated protein 5 (DAP5/eIF4G2/NAT1), a translation initiation factor mediating IRES-dependent translation. We found that the DAP5 knockdown from human ESCs (hESCs) resulted in persistence of pluripotent gene expression, delayed induction of differentiation-associated genes in different cell lineages, and defective embryoid body formation. The latter involved improper cellular organization, lack of cavitation, and enhanced mislocalized apoptosis. RNA sequencing of polysome-associated mRNAs identified candidates with reduced translation efficiency in DAP5-depleted hESCs. These were enriched in mitochondrial proteins involved in oxidative respiration, a pathway essential for differentiation, the significance of which was confirmed by the aberrant mitochondrial morphology and decreased oxidative respiratory activity in DAP5 knockdown cells. Further analysis identified the chromatin modifier HMGN3 as a cap-independent DAP5 translation target whose knockdown resulted in defective differentiation. Thus, DAP5-mediated translation of a specific set of proteins is critical for the transition from pluripotency to differentiation, highlighting the importance of cap-independent translation in stem cell fate decisions.


Asunto(s)
Diferenciación Celular/genética , Factor 4G Eucariótico de Iniciación/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Embrionarias Humanas/citología , Apoptosis/genética , Cuerpos Embrioides/patología , Factor 4G Eucariótico de Iniciación/genética , Técnicas de Silenciamiento del Gen , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Humanos , Células Madre Pluripotentes/fisiología
19.
Nat Commun ; 7: 11904, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27312851

RESUMEN

Death-associated protein kinase (DAPK) is a tumour suppressor. Here we show that DAPK also inhibits T helper 17 (Th17) and prevents Th17-mediated pathology in a mouse model of autoimmunity. We demonstrate that DAPK specifically downregulates hypoxia-inducible factor 1α (HIF-1α). In contrast to the predominant nuclear localization of HIF-1α in many cell types, HIF-1α is located in both the cytoplasm and nucleus in T cells, allowing for a cytosolic DAPK-HIF-1α interaction. DAPK also binds prolyl hydroxylase domain protein 2 (PHD2) and increases HIF-1α-PHD2 association. DAPK thereby promotes the proline hydroxylation and proteasome degradation of HIF-1α. Consequently, DAPK deficiency leads to excess HIF-1α accumulation, enhanced IL-17 expression and exacerbated experimental autoimmune encephalomyelitis. Additional knockout of HIF-1α restores the normal differentiation of Dapk(-/-) Th17 cells and prevents experimental autoimmune encephalomyelitis development. Our results reveal a mechanism involving DAPK-mediated degradation of cytoplasmic HIF-1α, and suggest that raising DAPK levels could be used for treatment of Th17-associated inflammatory diseases.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/genética , Encefalomielitis Autoinmune Experimental/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Células Th17/inmunología , Animales , Proteínas Quinasas Asociadas a Muerte Celular/deficiencia , Proteínas Quinasas Asociadas a Muerte Celular/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Hidroxilación , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Prolina Dioxigenasas del Factor Inducible por Hipoxia/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Células Jurkat , Ratones , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Toxina del Pertussis/administración & dosificación , Prolina/metabolismo , Complejo de la Endopetidasa Proteasomal , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células Th17/efectos de los fármacos , Células Th17/patología
20.
Nucleic Acids Res ; 43(7): 3764-75, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25779044

RESUMEN

Initiation is a highly regulated rate-limiting step of mRNA translation. During cap-dependent translation, the cap-binding protein eIF4E recruits the mRNA to the ribosome. Specific elements in the 5'UTR of some mRNAs referred to as Internal Ribosome Entry Sites (IRESes) allow direct association of the mRNA with the ribosome without the requirement for eIF4E. Cap-independent initiation permits translation of a subset of cellular and viral mRNAs under conditions wherein cap-dependent translation is inhibited, such as stress, mitosis and viral infection. DAP5 is an eIF4G homolog that has been proposed to regulate both cap-dependent and cap-independent translation. Herein, we demonstrate that DAP5 associates with eIF2ß and eIF4AI to stimulate IRES-dependent translation of cellular mRNAs. In contrast, DAP5 is dispensable for cap-dependent translation. These findings provide the first mechanistic insights into the function of DAP5 as a selective regulator of cap-independent translation.


Asunto(s)
Factor 2B Eucariótico de Iniciación/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Biosíntesis de Proteínas , Ribosomas/metabolismo , Células HEK293 , Humanos , Caperuzas de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...