Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Neuron ; 102(3): 602-620.e9, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30902550

RESUMEN

The rich functional diversity of the nervous system is founded in the specific connectivity of the underlying neural circuitry. Neurons are often preprogrammed to respond to multiple axon guidance signals because they use sequential guideposts along their pathways, but this necessitates a strict spatiotemporal regulation of intracellular signaling to ensure the cues are detected in the correct order. We performed a mouse mutagenesis screen and identified the Rho GTPase antagonist p190RhoGAP as a critical regulator of motor axon guidance. Rather than acting as a compulsory signal relay, p190RhoGAP uses a non-conventional GAP-independent mode to transiently suppress attraction to Netrin-1 while motor axons exit the spinal cord. Once in the periphery, a subset of axons requires p190RhoGAP-mediated inhibition of Rho signaling to target specific muscles. Thus, the multifunctional activity of p190RhoGAP emerges from its modular design. Our findings reveal a cell-intrinsic gate that filters conflicting signals, establishing temporal windows of signal detection.


Asunto(s)
Orientación del Axón/genética , Receptor DCC/metabolismo , Proteínas Activadoras de GTPasa/genética , Neuronas Motoras/metabolismo , Músculo Esquelético/inervación , Netrina-1/metabolismo , Proteínas Represoras/genética , Animales , Células del Asta Anterior/metabolismo , Ratones , Células Madre Embrionarias de Ratones , Mutación
2.
J Biol Chem ; 294(8): 2665-2677, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30593505

RESUMEN

Tyrosyl-DNA phosphodiesterase 2 (TDP2) is a multifunctional protein that has been implicated in a myriad of cellular pathways. Although most well-known for its phosphodiesterase activity removing stalled topoisomerase 2 from DNA, TDP2 has also been shown to interact with both survival and apoptotic mitogen-activated protein kinase (MAPK) signaling cascades. Moreover, it facilitates enterovirus replication and has been genetically linked to neurological disorders ranging from Parkinson's disease to dyslexia. To accurately evaluate TDP2 as a therapeutic target, we need to understand how TDP2 performs such a wide diversity of functions. Here, we use cancer cell lines modified with CRISPR/Cas9 or stably-expressed TDP2-targeted shRNA and transfection of various TDP2 mutants to show that its expression is regulated at the translational level via an internal ribosome entry site (IRES) that initiates translation at codon 54, the second in-frame methionine of the TDP2 coding sequence. We observed that this IRES drives expression of a shorter, N-terminally truncated isoform of TDP2, ΔN-TDP2, which omits a nuclear localization sequence. Additionally, we noted that ΔN-TDP2 retains phosphodiesterase activity and is protective against etoposide-induced cell death, but co-immunoprecipitates with fewer high-molecular-weight ubiquitinated peptide species, suggesting partial loss-of-function of TDP2's ubiquitin-association domain. In summary, our findings suggest the existence of an IRES in the 5' coding sequence of TDP2 that translationally regulates expression of an N-terminally truncated, cytoplasmic isoform of TDP2. These results shed light on the regulation of this multifunctional protein and may inform the design of therapies targeting TDP2 and associated pathways.


Asunto(s)
Empalme Alternativo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Sitios Internos de Entrada al Ribosoma/genética , Neoplasias/genética , Proteínas Nucleares/genética , Ribosomas/metabolismo , Factores de Transcripción/genética , Secuencia de Aminoácidos , Proteínas de Unión al ADN , Humanos , Neoplasias/enzimología , Neoplasias/patología , Iniciación de la Cadena Peptídica Traduccional , Hidrolasas Diéster Fosfóricas , Isoformas de Proteínas , Ribosomas/genética , Homología de Secuencia , Células Tumorales Cultivadas , Ubiquitina/metabolismo
3.
Mol Cell ; 70(5): 842-853.e7, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861157

RESUMEN

Heterochromatic repetitive satellite RNAs are extensively transcribed in a variety of human cancers, including BRCA1 mutant breast cancer. Aberrant expression of satellite RNAs in cultured cells induces the DNA damage response, activates cell cycle checkpoints, and causes defects in chromosome segregation. However, the mechanism by which satellite RNA expression leads to genomic instability is not well understood. Here we provide evidence that increased levels of satellite RNAs in mammary glands induce tumor formation in mice. Using mass spectrometry, we further show that genomic instability induced by satellite RNAs occurs through interactions with BRCA1-associated protein networks required for the stabilization of DNA replication forks. Additionally, de-stabilized replication forks likely promote the formation of RNA-DNA hybrids in cells expressing satellite RNAs. These studies lay the foundation for developing novel therapeutic strategies that block the effects of non-coding satellite RNAs in cancer cells.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Daño del ADN , Inestabilidad Genómica , Heterocromatina/genética , ARN Neoplásico/genética , Satélite de ARN/genética , Animales , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Heterocromatina/metabolismo , Humanos , Células MCF-7 , Ratones , Unión Proteica , ARN Neoplásico/metabolismo , Satélite de ARN/metabolismo , Carga Tumoral
4.
J Biol Chem ; 292(2): 611-628, 2017 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-27909058

RESUMEN

Glioblastomas (GBMs) are malignant brain tumors with a median survival of less than 18 months. Redundancy of signaling pathways represented within GBMs contributes to their therapeutic resistance. Exosomes are extracellular nanovesicles released from cells and present in human biofluids that represent a possible biomarker of tumor signaling state that could aid in personalized treatment. Herein, we demonstrate that mouse GBM cell-derived extracellular nanovesicles resembling exosomes from an H-RasV12 myr-Akt mouse model for GBM are enriched for intracellular signaling cascade proteins (GO: 0007242) and Ras protein signal transduction (GO: 0007265), and contain active Ras. Active Ras isolated from human and mouse GBM extracellular nanovesicles lysates using the Ras-binding domain of Raf also coprecipitates with ESCRT (endosomal sorting complex required for transport)-associated exosome proteins Vps4a and Alix. Although we initially hypothesized a role for active Ras protein signaling in exosome biogenesis, we found that GTP binding of K-Ras was dispensable for its packaging within extracellular nanovesicles and for the release of Alix. By contrast, farnesylation of K-Ras was required for its packaging within extracellular nanovesicles, yet expressing a K-Ras farnesylation mutant did not decrease the number of nanovesicles or the amount of Alix protein released per cell. Overall, these results emphasize the primary importance of membrane association in packaging of extracellular nanovesicle factors and indicate that screening nanovesicles within human fluids could provide insight into tissue origin and the wiring of signaling proteins at membranes to predict onset and behavior of cancer and other diseases linked to deregulated membrane signaling states.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Glioblastoma/metabolismo , Prenilación , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/patología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética , Quinasas raf/genética , Quinasas raf/metabolismo
5.
Cell ; 163(3): 583-93, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26496605

RESUMEN

LINE-1 retrotransposons are fast-evolving mobile genetic entities that play roles in gene regulation, pathological conditions, and evolution. Here, we show that the primate LINE-1 5'UTR contains a primate-specific open reading frame (ORF) in the antisense orientation that we named ORF0. The gene product of this ORF localizes to promyelocytic leukemia-adjacent nuclear bodies. ORF0 is present in more than 3,000 loci across human and chimpanzee genomes and has a promoter and a conserved strong Kozak sequence that supports translation. By virtue of containing two splice donor sites, ORF0 can also form fusion proteins with proximal exons. ORF0 transcripts are readily detected in induced pluripotent stem (iPS) cells from both primate species. Capped and polyadenylated ORF0 mRNAs are present in the cytoplasm, and endogenous ORF0 peptides are identified upon proteomic analysis. Finally, ORF0 enhances LINE-1 mobility. Taken together, these results suggest a role for ORF0 in retrotransposon-mediated diversity.


Asunto(s)
Pan troglodytes/genética , Retroelementos , Regiones no Traducidas 5' , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Citoplasma/genética , Humanos , Elementos de Nucleótido Esparcido Largo , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Sistemas de Lectura Abierta , Procesamiento Postranscripcional del ARN , ARN sin Sentido/genética , ARN Mensajero/química , ARN Mensajero/genética , Ribosomas/metabolismo , Alineación de Secuencia
6.
Cell ; 162(1): 198-210, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26140597

RESUMEN

Histidine phosphorylation (pHis) is well studied in bacteria; however, its role in mammalian signaling remains largely unexplored due to the lack of pHis-specific antibodies and the lability of the phosphoramidate (P-N) bond. Both imidazole nitrogens can be phosphorylated, forming 1-phosphohistidine (1-pHis) or 3-phosphohistidine (3-pHis). We have developed monoclonal antibodies (mAbs) that specifically recognize 1-pHis or 3-pHis; they do not cross-react with phosphotyrosine or the other pHis isomer. Assays based on the isomer-specific autophosphorylation of NME1 and phosphoglycerate mutase were used with immunoblotting and sequencing IgG variable domains to screen, select, and characterize anti-1-pHis and anti-3-pHis mAbs. Their sequence independence was determined by blotting synthetic peptide arrays, and they have been tested for immunofluorescence staining and immunoaffinity purification, leading to putative identification of pHis-containing proteins. These reagents should be broadly useful for identification of pHis substrates and functional study of pHis using a variety of immunological, proteomic, and biological assays.


Asunto(s)
Anticuerpos Monoclonales , Histidina/metabolismo , Animales , Centrosoma , Cromatografía Liquida , Células HeLa , Humanos , Modelos Químicos , Péptidos/análisis , Fosforilación , Polos del Huso , Espectrometría de Masas en Tándem
7.
Mol Cell Proteomics ; 13(7): 1659-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24797264

RESUMEN

Post-translational modification by SUMO is a highly conserved pathway in eukaryotes that plays very important regulatory roles in many cellular processes. Deregulation of the SUMO pathway contributes to the development and progression of many diseases including cancer. Therefore, identifying additional SUMO substrates and studying how their cellular and biological functions are regulated by sumoylation should provide new insights. Our studies showed that sumoylation activity was significant in Xenopus egg extracts, and that a high level of sumoylation was associated with sperm chromatin when SUMO was incubated with Xenopus egg extracts. By isolating SUMO-conjugated substrates using His-tagged SUMO1 or SUMO2 proteins under denaturing conditions, we identified 346 proteins by mass spectrometry analysis that were not present in control pull-downs. Among them, 167 proteins were identified from interphase egg extracts, 86 proteins from mitotic phase egg extracts, and 93 proteins from both. Thirty-three proteins were pulled down by SUMO1, 85 proteins by SUMO2, and 228 proteins by both. We validated the sumoylation of five candidates, CKB, ATXN10, BTF3, HABP4, and BZW1, by co-transfecting them along with SUMO in HEK293T cells. Gene ontology analysis showed that SUMO substrates identified in this study were involved in diverse biological processes. Additionally, SUMO substrates identified from different cell cycle stages or pulled down by different SUMO homologs were enriched for distinct cellular components and functional categories. Our results comprehensively profile the sumoylation occurring in the Xenopus egg extract system.


Asunto(s)
Cromatina/metabolismo , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Espermatozoides/metabolismo , Sumoilación/fisiología , Secuencia de Aminoácidos , Animales , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quimiocinas CC/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Datos de Secuencia Molecular , Factores Reguladores Miogénicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Óvulo/metabolismo , Factores de Transcripción/metabolismo , Xenopus
8.
Cell Rep ; 7(3): 722-34, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24794442

RESUMEN

Stem cells reside within specialized microenvironments, or niches, that control many aspects of stem cell behavior. Somatic hub cells in the Drosophila testis regulate the behavior of cyst stem cells (CySCs) and germline stem cells (GSCs) and are a primary component of the testis stem cell niche. The shutoff (shof) mutation, characterized by premature loss of GSCs and CySCs, was mapped to a locus encoding the evolutionarily conserved transcription factor Escargot (Esg). Hub cells depleted of Esg acquire CySC characteristics and differentiate as cyst cells, resulting in complete loss of hub cells and eventually CySCs and GSCs, similar to the shof mutant phenotype. We identified Esg-interacting proteins and demonstrate an interaction between Esg and the corepressor C-terminal binding protein (CtBP), which was also required for maintenance of hub cell fate. Our results indicate that niche cells can acquire stem cell properties upon removal of a single transcription factor in vivo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Nicho de Células Madre , Células Madre/citología , Testículo/citología , Alelos , Animales , Linaje de la Célula , Proteínas de Drosophila/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Células Madre/metabolismo , Factores de Transcripción/metabolismo
9.
Bioinformatics ; 30(15): 2208-9, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24681903

RESUMEN

MOTIVATION: We introduce Census 2, an update of a mass spectrometry data analysis tool for peptide/protein quantification. New features for analysis of isobaric labeling, such as Tandem Mass Tag (TMT) or Isobaric Tags for Relative and Absolute Quantification (iTRAQ), have been added in this version, including a reporter ion impurity correction, a reporter ion intensity threshold filter and an option for weighted normalization to correct mixing errors. TMT/iTRAQ analysis can be performed on experiments using HCD (High Energy Collision Dissociation) only, CID (Collision Induced Dissociation)/HCD (High Energy Collision Dissociation) dual scans or HCD triple-stage mass spectrometry data. To improve measurement accuracy, we implemented weighted normalization, multiple tandem spectral approach, impurity correction and dynamic intensity threshold features. AVAILABILITY AND IMPLEMENTATION: Census 2 supports multiple input file formats including MS1/MS2, DTASelect, mzXML and pepXML. It requires JAVA version 6 or later to run. Free download of Census 2 for academic users is available at http://fields.scripps.edu/census/index.php. CONTACT: jyates@scripps.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Espectrometría de Masas/métodos , Proteómica/métodos , Estadística como Asunto/métodos , Animales , Línea Celular , Marcaje Isotópico , Ratones , Péptidos/análisis , Péptidos/química , Proteínas/análisis , Proteínas/química
10.
DNA Repair (Amst) ; 15: 29-38, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24461736

RESUMEN

Faithful transmission of genetic material is essential for cell viability and organism health. The occurrence of DNA damage, due to either spontaneous events or environmental agents, threatens the integrity of the genome. The consequences of these insults, if allowed to perpetuate and accumulate over time, are mutations that can lead to the development of diseases such as cancer. Alkylation is a relevant DNA lesion produced endogenously as well as by exogenous agents including certain chemotherapeutics. We sought to better understand the cellular response to this form of DNA damage using mass spectrometry-based proteomics. For this purpose, we performed sub-cellular fractionation to monitor the effect of methyl methanesulfonate (MMS) treatment on protein localization to chromatin. The levels of over 500 proteins were increased in the chromatin-enriched nuclear lysate including histone chaperones. Levels of ubiquitin and subunits of the proteasome were also increased within this fraction, suggesting that ubiquitin-mediated degradation by the proteasome has an important role in the chromatin response to MMS treatment. Finally, the levels of some proteins were decreased within the chromatin-enriched lysate including components of the nuclear pore complex. Our spatial proteomics data demonstrate that many proteins that influence chromatin organization are regulated in response to MMS treatment, presumably to open the DNA to allow access by other DNA damage response proteins. To gain further insight into the cellular response to MMS-induced DNA damage, we also performed phosphorylation enrichment on total cell lysates to identify proteins regulated via post-translational modification. Phosphoproteomic analysis demonstrated that many nuclear phosphorylation events were decreased in response to MMS treatment. This reflected changes in protein kinase and/or phosphatase activity in response to DNA damage rather than changes in total protein abundance. Using these two mass spectrometry-based approaches, we have identified a novel set of MMS-responsive proteins that will expand our understanding of DNA damage signaling.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Reparación del ADN , Metilmetanosulfonato/toxicidad , Proteoma/metabolismo , Cromatina/metabolismo , Daño del ADN , Células HeLa , Humanos , Espectrometría de Masas , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional
11.
Mol Cell Biol ; 34(3): 452-63, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24277933

RESUMEN

The reversible nature of protein phosphorylation dictates that any protein kinase activity must be counteracted by protein phosphatase activity. How phosphatases target specific phosphoprotein substrates and reverse the action of kinases, however, is poorly understood in a biological context. We address this question by elucidating a novel function of the conserved PP4 family phosphatase Pph3-Psy2, the yeast counterpart of the mammalian PP4c-R3 complex, in the glucose-signaling pathway. Our studies show that Pph3-Psy2 specifically targets the glucose signal transducer protein Mth1 via direct binding of the EVH1 domain of the Psy2 regulatory subunit to the polyproline motif of Mth1. This activity is required for the timely dephosphorylation of the downstream transcriptional repressor Rgt1 upon glucose withdrawal, a critical event in the repression of HXT genes, which encode glucose transporters. Pph3-Psy2 dephosphorylates Mth1, an Rgt1 associated corepressor, but does not dephosphorylate Rgt1 at sites associated with inactivation, in vitro. We show that Pph3-Psy2 phosphatase antagonizes Mth1 phosphorylation by protein kinase A (PKA), the major protein kinase activated in response to glucose, in vitro and regulates Mth1 function via putative PKA phosphorylation sites in vivo. We conclude that the Pph3-Psy2 phosphatase modulates Mth1 activity to facilitate precise regulation of HXT gene expression by glucose.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Immunoblotting , Mutación , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , Fosforilación/efectos de los fármacos , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Técnicas del Sistema de Dos Híbridos
12.
Mol Cell ; 50(5): 699-710, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23685072

RESUMEN

Oncogene-induced senescence is an important tumor-suppressing defense mechanism. However, relatively little is known about the signaling pathway mediating the senescence response. Here, we demonstrate that a multifunctional acetyltransferase, Tip60, plays an essential role in oncogenic ras-induced senescence. Further investigation reveals a cascade of posttranslational modifications involving p38, Tip60, and PRAK, three proteins that are essential for ras-induced senescence. Upon activation by ras, p38 induces the acetyltransferase activity of Tip60 through phosphorylation of Thr158; activated Tip60 in turn directly interacts with and induces the protein kinase activity of PRAK through acetylation of K364 in a manner that depends on phosphorylation of both Tip60 and PRAK by p38. These posttranslational modifications are critical for the prosenescent function of Tip60 and PRAK, respectively. These results have defined a signaling pathway that mediates oncogene-induced senescence, and identified posttranslational modifications that regulate the enzymatic activity and biological functions of Tip60 and PRAK.


Asunto(s)
Senescencia Celular/genética , Genes ras , Histona Acetiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Acetilación , Línea Celular , Histona Acetiltransferasas/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lisina Acetiltransferasa 5 , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Treonina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
13.
J Biol Chem ; 287(52): 43984-94, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23115235

RESUMEN

Ubiquitination plays an important role in the DNA damage response. We identified a novel interaction of the E3 ubiquitin ligase RNF8 with Nbs1, a key regulator of DNA double-strand break (DSB) repair. We found that Nbs1 is ubiquitinated both before and after DNA damage and is a direct ubiquitination substrate of RNF8. We also identified key residues on Nbs1 that are ubiquitinated by RNF8. By using laser microirradiation and live-cell imaging, we observed that RNF8 and its ubiquitination activity are important for promoting optimal binding of Nbs1 to DSB-containing chromatin. We also demonstrated that RNF8-mediated ubiquitination of Nbs1 contributes to the efficient and stable binding of Nbs1 to DSBs and is important for HR-mediated DSB repair. Taken together, these studies suggest that Nbs1 is one important target of RNF8 to regulate DNA DSB repair.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Recombinación Homóloga/fisiología , Proteínas Nucleares/metabolismo , Ubiquitinación/fisiología , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/genética , Recombinación Homóloga/efectos de la radiación , Humanos , Rayos Láser/efectos adversos , Proteínas Nucleares/genética , Ubiquitina-Proteína Ligasas , Ubiquitinación/efectos de la radiación
14.
J Biol Chem ; 287(35): 29610-9, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22730331

RESUMEN

Protein modification by SUMO and ubiquitin critically impacts genome stability via effectors that "read" their signals using SUMO interaction motifs or ubiquitin binding domains, respectively. A novel mixed SUMO and ubiquitin signal is generated by the SUMO-targeted ubiquitin ligase (STUbL), which ubiquitylates SUMO conjugates. Herein, we determine that the "ubiquitin-selective" segregase Cdc48-Ufd1-Npl4 also binds SUMO via a SUMO interaction motif in Ufd1 and can thus act as a selective receptor for STUbL targets. Indeed, we define key cooperative DNA repair functions for Cdc48-Ufd1-Npl4 and STUbL, thereby revealing a new signaling mechanism involving dual recruitment by SUMO and ubiquitin for Cdc48-Ufd1-Npl4 functions in maintaining genome stability.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Genómica/fisiología , Proteína SUMO-1/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/fisiología , Adenosina Trifosfatasas/genética , Secuencias de Aminoácidos , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Reparación del ADN/fisiología , ADN de Hongos/genética , ADN de Hongos/metabolismo , Unión Proteica , Proteína SUMO-1/genética , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Transducción de Señal/fisiología , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética , Proteína que Contiene Valosina
15.
J Biol Chem ; 285(48): 37733-40, 2010 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-20870719

RESUMEN

The retinoblastoma tumor suppressor (RB) is a central cell cycle regulator and tumor suppressor. RB cellular functions are known to be regulated by a diversity of post-translational modifications such as phosphorylation and acetylation, raising the possibility that RB may also be methylated in cells. Here we demonstrate that RB can be methylated by SMYD2 at lysine 860, a highly conserved and novel site of modification. This methylation event occurs in vitro and in cells, and it is regulated during cell cycle progression, cellular differentiation, and in response to DNA damage. Furthermore, we show that RB monomethylation at lysine 860 provides a direct binding site for the methyl-binding domain of the transcriptional repressor L3MBTL1. These results support the idea that a code of post-translational modifications exists for RB and helps guide its functions in mammalian cells.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de Retinoblastoma/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular Tumoral , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/genética , Alineación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
Mol Cell ; 35(4): 442-53, 2009 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-19716789

RESUMEN

ATR and Chk1 are two key protein kinases in the replication checkpoint. Activation of ATR-Chk1 has been extensively investigated, but checkpoint termination and replication fork restart are less well understood. Here, we report that DNA damage not only activates Chk1, but also exposes a degron-like region at the carboxyl terminus of Chk1 to an Fbx6-containing SCF (Skp1-Cul1-F box) E3 ligase, which mediates the ubiquitination and degradation of Chk1 and, in turn, terminates the checkpoint. The protein levels of Chk1 and Fbx6 showed an inverse correlation in both cultured cancer cells and in human breast tumor tissues. Further, we show that low levels of Fbx6 and consequent impairment of replication stress-induced Chk1 degradation are associated with cancer cell resistance to the chemotherapeutic agent, camptothecin. We propose that Fbx6-dependent Chk1 degradation contributes to S phase checkpoint termination and that a defect in this mechanism might increase tumor cell resistance to certain anticancer drugs.


Asunto(s)
Daño del ADN , Replicación del ADN , Neoplasias/enzimología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Ligasas SKP Cullina F-box/metabolismo , Estrés Fisiológico , Antineoplásicos Fitogénicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Camptotecina/farmacología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Activación Enzimática , Humanos , Lisina , Neoplasias/genética , Neoplasias/patología , Fosforilación , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Ligasas SKP Cullina F-box/genética , Factores de Tiempo , Ubiquitinación
17.
Cancer Cell ; 11(2): 175-89, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17292828

RESUMEN

In response to DNA damage, eukaryotic cells activate ATM-Chk2 and/or ATR-Chk1 to arrest the cell cycle and initiate DNA repair. We show that, in the absence of p53, cells depend on a third cell-cycle checkpoint pathway involving p38MAPK/MK2 for cell-cycle arrest and survival after DNA damage. MK2 depletion in p53-deficient cells, but not in p53 wild-type cells, caused abrogation of the Cdc25A-mediated S phase checkpoint after cisplatin exposure and loss of the Cdc25B-mediated G2/M checkpoint following doxorubicin treatment, resulting in mitotic catastrophe and pronounced regression of murine tumors in vivo. We show that the Chk1 inhibitor UCN-01 also potently inhibits MK2, suggesting that its clinical efficacy results from the simultaneous disruption of two critical checkpoint pathways in p53-defective cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Supervivencia Celular , Células Cultivadas , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Doxorrubicina/farmacología , Fase G2/efectos de los fármacos , Fase G2/efectos de la radiación , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Ratones Desnudos , Mitosis/efectos de los fármacos , Mitosis/efectos de la radiación , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Quinasas/genética , Fase S/efectos de los fármacos , Fase S/efectos de la radiación , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Rayos Ultravioleta , Fosfatasas cdc25/metabolismo
18.
Genes Dev ; 18(12): 1413-22, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15175242

RESUMEN

Tumor development is dependent upon the inactivation of two key tumor-suppressor networks, p16(Ink4a)-cycD/cdk4-pRB-E2F and p19(Arf)-mdm2-p53, that regulate cellular proliferation and the tumor surveillance response. These networks are known to intersect with one another, but the mechanisms are poorly understood. Here, we show that E2F directly participates in the transcriptional control of Arf in both normal and transformed cells. This occurs in a manner that is significantly different from the regulation of classic E2F-responsive targets. In wild-type mouse embryonic fibroblasts (MEFs), the Arf promoter is occupied by E2F3 and not other E2F family members. In quiescent cells, this role is largely fulfilled by E2F3b, an E2F3 isoform whose function was previously undetermined. E2f3 loss is sufficient to derepress Arf, triggering activation of p53 and expression of p21(Cip1). Thus, E2F3 is a key repressor of the p19(Arf)-p53 pathway in normal cells. Consistent with this notion, Arf mutation suppresses the activation of p53 and p21(Cip1) in E2f3-deficient MEFs. Arf loss also rescues the known cell cycle re-entry defect of E2f3(-/-) cells, and this correlates with restoration of appropriate activation of classic E2F-responsive genes. Our data also demonstrate a direct role for E2F in the oncogenic activation of Arf. Specifically, we observe recruitment of the endogenous activating E2Fs, E2F1, and E2F3a, to the Arf promoter. Thus, distinct E2F complexes directly contribute to the normal repression and oncogenic activation of Arf. We propose that monitoring of E2F levels and/or activity is a key component of Arf's ability to respond to inappropriate, but not normal cellular proliferation.


Asunto(s)
Ciclo Celular , Regulación hacia Abajo , Factores de Transcripción/fisiología , Proteína p14ARF Supresora de Tumor/genética , Animales , Línea Celular Transformada , Transformación Celular Neoplásica , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Factor de Transcripción E2F3 , Fibroblastos , Cinética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Factores de Transcripción/genética , Transcripción Genética , Transfección , Proteína p14ARF Supresora de Tumor/biosíntesis , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
19.
Mol Cell ; 11(4): 905-14, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12718877

RESUMEN

Myc and E2f1 promote cell cycle progression, but overexpression of either can trigger p53-dependent apoptosis. Mice expressing an Emu-Myc transgene in B lymphocytes develop lymphomas, the majority of which sustain mutations of either the Arf or p53 tumor suppressors. Emu-Myc transgenic mice lacking one or both E2f1 alleles exhibited a slower onset of lymphoma development associated with increased expression of the cyclin-dependent kinase inhibitor p27(Kip1) and a reduced S phase fraction in precancerous B cells. In contrast, Myc-induced apoptosis and the frequency of Arf and p53 mutations in lymphomas were unaffected by E2f1 loss. Therefore, Myc does not require E2f1 to induce Arf, p53, or apoptosis in B cells, but depends upon E2f1 to accelerate cell cycle progression and downregulate p27(Kip1).


Asunto(s)
Apoptosis/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN , Linfoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/deficiencia , Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Femenino , Genotipo , Linfoma/genética , Linfoma/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Proteínas Proto-Oncogénicas c-myc/genética , Fase S/genética , Factores de Transcripción/genética , Transgenes/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA