Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Circ Res ; 126(10): 1379-1393, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32175818

RESUMEN

RATIONALE: Noonan syndrome (NS) is one of the most frequent genetic disorders. Bleeding problems are among the most common, yet poorly defined complications associated with NS. A lack of consensus on the management of bleeding complications in patients with NS indicates an urgent need for new therapeutic approaches. OBJECTIVE: Bleeding disorders have recently been described in patients with NS harboring mutations of LZTR1 (leucine zipper-like transcription regulator 1), an adaptor for CUL3 (CULLIN3) ubiquitin ligase complex. Here, we assessed the pathobiology of LZTR1-mediated bleeding disorders. METHODS AND RESULTS: Whole-body and vascular specific knockout of Lztr1 results in perinatal lethality due to cardiovascular dysfunction. Lztr1 deletion in blood vessels of adult mice leads to abnormal vascular leakage. We found that defective adherent and tight junctions in Lztr1-depleted endothelial cells are caused by dysregulation of vesicular trafficking. LZTR1 affects the dynamics of fusion and fission of recycling endosomes by controlling ubiquitination of the ESCRT-III (endosomal sorting complex required for transport III) component CHMP1B (charged multivesicular protein 1B), whereas NS-associated LZTR1 mutations diminish CHMP1B ubiquitination. LZTR1-mediated dysregulation of CHMP1B ubiquitination triggers endosomal accumulation and subsequent activation of VEGFR2 (vascular endothelial growth factor receptor 2) and decreases blood levels of soluble VEGFR2 in Lztr1 haploinsufficient mice. Inhibition of VEGFR2 activity by cediranib rescues vascular abnormalities observed in Lztr1 knockout mice Conclusions: Lztr1 deletion phenotypically overlaps with bleeding diathesis observed in patients with NS. ELISA screening of soluble VEGFR2 in the blood of LZTR1-mutated patients with NS may predict both the severity of NS phenotypes and potential responders to anti-VEGF therapy. VEGFR inhibitors could be beneficial for the treatment of bleeding disorders in patients with NS.


Asunto(s)
Vasos Sanguíneos/metabolismo , Endosomas/metabolismo , Células Endoteliales/metabolismo , Hemorragia/metabolismo , Síndrome de Noonan/metabolismo , Factores de Transcripción/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Malformaciones Vasculares/metabolismo , Animales , Vasos Sanguíneos/anomalías , Vasos Sanguíneos/efectos de los fármacos , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Modelos Animales de Enfermedad , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Endosomas/genética , Endosomas/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Haploinsuficiencia , Células HeLa , Hemorragia/genética , Hemorragia/patología , Hemorragia/prevención & control , Humanos , Linfocinas/genética , Linfocinas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Síndrome de Noonan/tratamiento farmacológico , Síndrome de Noonan/genética , Síndrome de Noonan/patología , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Quinazolinas/farmacología , Transducción de Señal , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Ubiquitinación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Malformaciones Vasculares/tratamiento farmacológico , Malformaciones Vasculares/genética , Malformaciones Vasculares/patología
2.
Mol Cell ; 54(6): 932-945, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24857551

RESUMEN

Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.


Asunto(s)
Fase G1/genética , Fase G2/genética , Proteína Oncogénica p21(ras)/genética , Proteína Fosfatasa 2/genética , Fase de Descanso del Ciclo Celular/fisiología , Línea Celular , Ciclina E/biosíntesis , Humanos , Células MCF-7 , Mitosis/genética , Subunidades de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal/genética
3.
Nucleic Acids Res ; 48(5): 2502-2517, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-31956895

RESUMEN

Dysregulated splicing is a common event in cancer even in the absence of mutations in the core splicing machinery. The aberrant long non-coding transcriptome constitutes an uncharacterized level of regulation of post-transcriptional events in cancer. Here, we found that the stress-induced long non-coding RNA (lncRNA), LINC02657 or LASTR (lncRNA associated with SART3 regulation of splicing), is upregulated in hypoxic breast cancer and is essential for the growth of LASTR-positive triple-negative breast tumors. LASTR is upregulated in several types of epithelial cancers due to the activation of the stress-induced JNK/c-JUN pathway. Using a mass-spectrometry based approach, we identified the RNA-splicing factor SART3 as a LASTR-interacting partner. We found that LASTR promotes splicing efficiency by controlling SART3 association with the U4 and U6 small nuclear ribonucleoproteins (snRNP) during spliceosome recycling. Intron retention induced by LASTR depletion downregulates expression of essential genes, ultimately decreasing the fitness of cancer cells.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias/genética , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Estrés Fisiológico , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Genes Esenciales , Humanos , Intrones/genética , Sistema de Señalización de MAP Quinasas , Ratones Desnudos , Empalme del ARN/genética , ARN Largo no Codificante/genética , Regulación hacia Arriba/genética
4.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344852

RESUMEN

Ubiquitination is a versatile and dynamic post-translational modification in which single ubiquitin molecules or polyubiquitin chains are attached to target proteins, giving rise to mono- or poly-ubiquitination, respectively. The majority of research in the ubiquitin field focused on degradative polyubiquitination, whereas more recent studies uncovered the role of single ubiquitin modification in important physiological processes. Monoubiquitination can modulate the stability, subcellular localization, binding properties, and activity of the target proteins. Understanding the function of monoubiquitination in normal physiology and pathology has important therapeutic implications, as alterations in the monoubiquitin pathway are found in a broad range of genetic diseases. This review highlights a link between monoubiquitin signaling and the pathogenesis of genetic disorders.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Enfermedades Genéticas Congénitas/metabolismo , Procesamiento Proteico-Postraduccional , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina/metabolismo , Proteínas Ubiquitinadas/biosíntesis , Ubiquitinación , Enfermedades Genéticas Congénitas/genética , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Transporte de Proteínas , Proteolisis , Fracciones Subcelulares/metabolismo , Especificidad por Sustrato
6.
Nat Med ; 11(12): 1306-13, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16286925

RESUMEN

It is widely accepted that the p53 tumor suppressor restricts abnormal cells by induction of growth arrest or by triggering apoptosis. Here we show that, in addition, p53 protects the genome from oxidation by reactive oxygen species (ROS), a major cause of DNA damage and genetic instability. In the absence of severe stresses, relatively low levels of p53 are sufficient for upregulation of several genes with antioxidant products, which is associated with a decrease in intracellular ROS. Downregulation of p53 results in excessive oxidation of DNA, increased mutation rate and karyotype instability, which are prevented by incubation with the antioxidant N-acetylcysteine (NAC). Dietary supplementation with NAC prevented frequent lymphomas characteristic of Trp53-knockout mice, and slowed the growth of lung cancer xenografts deficient in p53. Our results provide a new paradigm for a nonrestrictive tumor suppressor function of p53 and highlight the potential importance of antioxidants in the prophylaxis and treatment of cancer.


Asunto(s)
Apoptosis/fisiología , Daño del ADN , Regulación de la Expresión Génica/fisiología , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Acetilcisteína/farmacología , Animales , Northern Blotting , Western Blotting , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Cartilla de ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Vectores Genéticos , Inestabilidad Genómica/efectos de los fármacos , Humanos , Cariotipificación , Lentivirus , Ratones , Mutagénesis , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Proteína p53 Supresora de Tumor/genética
7.
Curr Opin Struct Biol ; 73: 102333, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35176591

RESUMEN

The conjugation of a single ubiquitin or monoubiquitination acts as a versatile signal that can have both degradative and non-degradative functions. The latter is of particular interest as emerging evidence indicates that ubiquitin-driven alterations of the protein interaction landscape play a key role in multiple signaling pathways. Whereas early studies were focused on how monoubiquitination alters the interactions of proteins containing ubiquitin-binding domains, more recent reports demonstrate that ubiquitin conjugation can also affect the binding mode by changing the surface of the ubiquitinated substrate. Furthermore, monoubiquitination modulates the interactions with other macromolecules, such as DNA or lipids, underscoring the diverse role of monoubiquitination in cellular processes. In this review, we discussed how monoubiquitination achieves its function by modulating the interaction landscape.


Asunto(s)
Mapas de Interacción de Proteínas , Ubiquitina , Unión Proteica , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
8.
Mol Cancer Res ; 19(6): 979-990, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33619226

RESUMEN

Loss of chromosome 9p21 is observed in one-thirds of clear-cell renal cell carcinoma (ccRCC) and is associated with poorer patient survival. Unexpectedly, 9p21 LOH does not lead to decreased expression of the 9p21 tumor suppressor genes, CDKN2A and CDKN2B, suggesting alternative mechanisms of 9p-mediated tumorigenesis. Concordantly, CRISPR-mediated 9p21 deletion promotes growth of immortalized human embryonic kidney epithelial cells independently of the CDKN2A/B pathway inactivation. The 9p21 locus has a highly accessible chromatin structure, suggesting that 9p21 loss might contribute to kidney cancer progression by dysregulating genes distal to the 9p21 locus. We identified several 9p21 regulatory hubs by assessing which of the 9p21-interacting genes are dysregulated in 9p21-deleted kidney cells and ccRCCs. By focusing on the analysis of the homeobox gene 13 (HOXB13) locus, we found that 9p21 loss relieves the HOXB13 locus, decreasing HOXB13 methylation and promoting its expression. Upregulation of HOXB13 facilitates cell growth and is associated with poorer survival of patients with ccRCC. IMPLICATIONS: The results of our study propose a novel tumor suppressive mechanism on the basis of coordinated expression of physically associated genes, providing a better understanding of the role of chromosomal deletions in cancer.


Asunto(s)
Carcinoma de Células Renales/genética , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Proteínas de Homeodominio/genética , Neoplasias Renales/genética , Regulación hacia Arriba , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Pérdida de Heterocigocidad , ARN Largo no Codificante/genética , RNA-Seq/métodos
9.
Sci Rep ; 11(1): 3176, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542435

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, lacking effective therapy. Many TNBCs show remarkable response to carboplatin-based chemotherapy, but often develop resistance over time. With increasing use of carboplatin in the clinic, there is a pressing need to identify vulnerabilities of carboplatin-resistant tumors. In this study, we generated carboplatin-resistant TNBC MDA-MB-468 cell line and patient derived TNBC xenograft models. Mass spectrometry-based proteome profiling demonstrated that carboplatin resistance in TNBC is linked to drastic metabolism rewiring and upregulation of anti-oxidative response that supports cell replication by maintaining low levels of DNA damage in the presence of carboplatin. Carboplatin-resistant cells also exhibited dysregulation of the mitotic checkpoint. A kinome shRNA screen revealed that carboplatin-resistant cells are vulnerable to the depletion of the mitotic checkpoint regulators, whereas the checkpoint kinases CHEK1 and WEE1 are indispensable for the survival of carboplatin-resistant cells in the presence of carboplatin. We confirmed that pharmacological inhibition of CHEK1 by prexasertib in the presence of carboplatin is well tolerated by mice and suppresses the growth of carboplatin-resistant TNBC xenografts. Thus, abrogation of the mitotic checkpoint by CHEK1 inhibition re-sensitizes carboplatin-resistant TNBCs to carboplatin and represents a potential strategy for the treatment of carboplatin-resistant TNBCs.


Asunto(s)
Carboplatino/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas Tirosina Quinasas/genética , Pirazinas/farmacología , Pirazoles/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Daño del ADN , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas de Neoplasias/clasificación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteoma/clasificación , Proteoma/genética , Proteoma/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Res ; 81(16): 4218-4229, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34215617

RESUMEN

Meningiomas are the most common benign brain tumors. Mutations of the E3 ubiquitin ligase TRAF7 occur in 25% of meningiomas and commonly cooccur with mutations in KLF4, yet the functional link between TRAF7 and KLF4 mutations remains unclear. By generating an in vitro meningioma model derived from primary meningeal cells, we elucidated the cooperative interactions that promote meningioma development. By integrating TRAF7-driven ubiquitinome and proteome alterations in meningeal cells and the TRAF7 interactome, we identified TRAF7 as a proteostatic regulator of RAS-related small GTPases. Meningioma-associated TRAF7 mutations disrupted either its catalytic activity or its interaction with RAS GTPases. TRAF7 loss in meningeal cells altered actin dynamics and promoted anchorage-independent growth by inducing CDC42 and RAS signaling. TRAF deficiency-driven activation of the RAS/MAPK pathway promoted KLF4-dependent transcription that led to upregulation of the tumor-suppressive Semaphorin pathway, a negative regulator of small GTPases. KLF4 loss of function disrupted this negative feedback loop and enhanced mutant TRAF7-mediated cell transformation. Overall, this study provides new mechanistic insights into meningioma development, which could lead to novel treatment strategies. SIGNIFICANCE: The intricate molecular cross-talk between the ubiquitin ligase TRAF7 and the transcription factor KLF4 provides a first step toward the identification of new therapies for patients with meningioma.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación Neoplásica de la Expresión Génica , Meningioma/genética , Mutación , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Proteínas ras/genética , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Biología Computacional , Células HEK293 , Humanos , Factor 4 Similar a Kruppel/genética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteoma , Semaforinas/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Activación Transcripcional , Ubiquitina/química , Proteína de Unión al GTP cdc42/genética , Proteínas ras/metabolismo
11.
PLoS Biol ; 5(8): e202, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17608567

RESUMEN

The SV40 small t antigen (ST) is a potent oncoprotein that perturbs the function of protein phosphatase 2A (PP2A). ST directly interacts with the PP2A scaffolding A subunit and alters PP2A activity by displacing regulatory B subunits from the A subunit. We have determined the crystal structure of full-length ST in complex with PP2A A subunit at 3.1 A resolution. ST consists of an N-terminal J domain and a C-terminal unique domain that contains two zinc-binding motifs. Both the J domain and second zinc-binding motif interact with the intra-HEAT-repeat loops of HEAT repeats 3-7 of the A subunit, which overlaps with the binding site of the PP2A B56 subunit. Intriguingly, the first zinc-binding motif is in a position that may allow it to directly interact with and inhibit the phosphatase activity of the PP2A catalytic C subunit. These observations provide a structural basis for understanding the oncogenic functions of ST.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Conformación Proteica , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/química , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Virus 40 de los Simios/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Virales de Tumores/química , Antígenos Virales de Tumores/genética , Sitios de Unión , Cristalografía por Rayos X , Análisis Mutacional de ADN , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína/metabolismo , Alineación de Secuencia
12.
Trends Cancer ; 6(5): 369-379, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32249186

RESUMEN

Decades ago, Rap1, a small GTPase very similar to Ras, was observed to suppress oncogenic Ras phenotype, reverting its transformation. The proposed reason, persisting since, has been competition between Ras and Rap1 for a common target. Yet, none was found. There was also Rap1's puzzling suppression of Raf-1 versus activation of BRAF. Reemerging interest in Rap1 envisages capturing its Ras suppression action by inhibitors. Here, we review the literature and resolve the enigma. In vivo oncogenic Ras exists in isoform-distinct nanoclusters. The presence of Rap1 within the nanoclusters reduces the number of the clustered oncogenic Ras molecules, thus suppressing Raf-1 activation and mitogen-activated protein kinase (MAPK) signaling. Nanoclustering suggests that Rap1 suppression is Ras isoform dependent. Altogether, a potent Rap1-like inhibitor appears unlikely.


Asunto(s)
Neoplasias/patología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Cristalografía , Humanos , Sistema de Señalización de MAP Quinasas , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas B-raf/ultraestructura , Proteínas Proto-Oncogénicas c-raf/ultraestructura , Proteínas Proto-Oncogénicas p21(ras)/ultraestructura , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/ultraestructura
15.
EMBO Mol Med ; 8(3): 288-303, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26881969

RESUMEN

Activation of the RAS oncogenic pathway, frequently ensuing from mutations in RAS genes, is a common event in human cancer. Recent reports demonstrate that reversible ubiquitination of RAS GTPases dramatically affects their activity, suggesting that enzymes involved in regulating RAS ubiquitination may contribute to malignant transformation. Here, we identified the de-ubiquitinase OTUB1 as a negative regulator of RAS mono- and di-ubiquitination. OTUB1 inhibits RAS ubiquitination independently of its catalytic activity resulting in sequestration of RAS on the plasma membrane. OTUB1 promotes RAS activation and tumorigenesis in wild-type RAS cells. An increase of OTUB1 expression is commonly observed in non-small-cell lung carcinomas harboring wild-type KRAS and is associated with increased levels of ERK1/2 phosphorylation, high Ki67 score, and poorer patient survival. Our results strongly indicate that dysregulation of RAS ubiquitination represents an alternative mechanism of RAS activation during lung cancer development.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/fisiopatología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Línea Celular Tumoral , Enzimas Desubicuitinizantes , Modelos Animales de Enfermedad , Humanos , Ratones Desnudos , Ubiquitinación
16.
Cancer Cell ; 29(5): 751-766, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27165746

RESUMEN

Large-scale heterozygous deletions are a hallmark of cancer genomes. The concomitant loss of multiple genes creates vulnerabilities that are impossible to reveal through the study of individual genes. To delineate the functional outcome of chromosome 8p loss of heterozygosity (LOH), a common aberration in breast cancer, we modeled 8p LOH using TALEN-based genomic engineering. 8p LOH alters fatty acid and ceramide metabolism. The shift in lipid metabolism triggers invasiveness and confers tumor growth under stress conditions due to increased autophagy. The resistance of 8p-deleted cells to chemotherapeutic drugs concurs with poorer survival rates of breast cancer patients harboring an 8p LOH. The autophagy dependency of 8p-deleted cells provides the rational basis for treatment of 8p LOH tumors with autophagy inhibitors.


Asunto(s)
Neoplasias de la Mama/genética , Deleción Cromosómica , Cromosomas Humanos Par 8/genética , Metabolismo de los Lípidos/genética , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hibridación Fluorescente in Situ/métodos , Estimación de Kaplan-Meier , Metabolismo de los Lípidos/efectos de los fármacos , Análisis Multivariante , Pronóstico , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
17.
Nat Med ; 22(8): 861-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27376578

RESUMEN

In a search for mediators of the p53 tumor suppressor pathway, which induces pleiotropic and often antagonistic cellular responses, we identified the long noncoding RNA (lncRNA) NEAT1. NEAT1 is an essential architectural component of paraspeckle nuclear bodies, whose pathophysiological relevance remains unclear. Activation of p53, pharmacologically or by oncogene-induced replication stress, stimulated the formation of paraspeckles in mouse and human cells. Silencing Neat1 expression in mice, which prevents paraspeckle formation, sensitized preneoplastic cells to DNA-damage-induced cell death and impaired skin tumorigenesis. We provide mechanistic evidence that NEAT1 promotes ATR signaling in response to replication stress and is thereby engaged in a negative feedback loop that attenuates oncogene-dependent activation of p53. NEAT1 targeting in established human cancer cell lines induced synthetic lethality with genotoxic chemotherapeutics, including PARP inhibitors, and nongenotoxic activation of p53. This study establishes a key genetic link between NEAT1 paraspeckles, p53 biology and tumorigenesis and identifies NEAT1 as a promising target to enhance sensitivity of cancer cells to both chemotherapy and p53 reactivation therapy.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica/genética , ARN Largo no Codificante/genética , Proteína p53 Supresora de Tumor/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular Tumoral , Daño del ADN , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Células MCF-7 , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Lesiones Precancerosas/genética , Modelos de Riesgos Proporcionales , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Neoplasias Cutáneas/genética , Análisis de Supervivencia
18.
Nat Cell Biol ; 15(10): 1220-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24056301

RESUMEN

The RAS-like GTPase RALB mediates cellular responses to nutrient availability or viral infection by respectively engaging two components of the exocyst complex, EXO84 and SEC5. RALB employs SEC5 to trigger innate immunity signalling, whereas RALB-EXO84 interaction induces autophagocytosis. How this differential interaction is achieved molecularly by the RAL GTPase remains unknown. We found that whereas GTP binding turns on RALB activity, ubiquitylation of RALB at Lys 47 tunes its activity towards a particular effector. Specifically, ubiquitylation at Lys 47 sterically inhibits RALB binding to EXO84, while facilitating its interaction with SEC5. Double-stranded RNA promotes RALB ubiquitylation and SEC5-TBK1 complex formation. In contrast, nutrient starvation induces RALB deubiquitylation by accumulation and relocalization of the deubiquitylase USP33 to RALB-positive vesicles. Deubiquitylated RALB promotes the assembly of the RALB-EXO84-beclin-1 complexes driving autophagosome formation. Thus, ubiquitylation within the effector-binding domain provides the switch for the dual functions of RALB in autophagy and innate immune responses.


Asunto(s)
Autofagia/fisiología , Inmunidad Innata/fisiología , Ubiquitina Tiolesterasa/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Autofagia/genética , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/genética , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Transducción de Señal , Ubiquitina Tiolesterasa/genética , Ubiquitinación , Proteínas de Unión al GTP ral/química , Proteínas de Unión al GTP ral/genética
19.
Anticancer Agents Med Chem ; 11(1): 38-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21288198

RESUMEN

The kinase oncogenes are well-characterized drivers of cancer development, and several targeted therapies focused on both specific and selectively nonselective kinase inhibitors have now been approved for clinical use. In contrast, much less is known about the role of protein phosphatases, although modulation of their activities might form the foundation for an effective anti-cancer approach. The serine-threonine protein phosphatase 2A (PP2A) is implicated in the regulation of numerous signaling pathways and may function as a tumor suppressor. Recently pharmacological modulation of PP2A activity has been showed to have a potent anti-tumor activity in both in vitro and in vivo cancer models. These studies implicate PP2A as a promising therapeutic target for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Proteína Fosfatasa 2/antagonistas & inhibidores , Animales , Antineoplásicos/química , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Neoplasias/metabolismo , Proteína Fosfatasa 2/metabolismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA