Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Mol Cell ; 83(12): 1983-2002.e11, 2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37295433

RESUMEN

The evolutionarily conserved minor spliceosome (MiS) is required for protein expression of ∼714 minor intron-containing genes (MIGs) crucial for cell-cycle regulation, DNA repair, and MAP-kinase signaling. We explored the role of MIGs and MiS in cancer, taking prostate cancer (PCa) as an exemplar. Both androgen receptor signaling and elevated levels of U6atac, a MiS small nuclear RNA, regulate MiS activity, which is highest in advanced metastatic PCa. siU6atac-mediated MiS inhibition in PCa in vitro model systems resulted in aberrant minor intron splicing leading to cell-cycle G1 arrest. Small interfering RNA knocking down U6atac was ∼50% more efficient in lowering tumor burden in models of advanced therapy-resistant PCa compared with standard antiandrogen therapy. In lethal PCa, siU6atac disrupted the splicing of a crucial lineage dependency factor, the RE1-silencing factor (REST). Taken together, we have nominated MiS as a vulnerability for lethal PCa and potentially other cancers.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Masculino , Humanos , Intrones/genética , Neoplasias de la Próstata/metabolismo , Empalme del ARN/genética , Empalmosomas/metabolismo , Transducción de Señal , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Neoplasias de la Próstata Resistentes a la Castración/genética
2.
Am J Hum Genet ; 106(3): 405-411, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109420

RESUMEN

Recurrent somatic variants in SPOP are cancer specific; endometrial and prostate cancers result from gain-of-function and dominant-negative effects toward BET proteins, respectively. By using clinical exome sequencing, we identified six de novo pathogenic missense variants in SPOP in seven individuals with developmental delay and/or intellectual disability, facial dysmorphisms, and congenital anomalies. Two individuals shared craniofacial dysmorphisms, including congenital microcephaly, that were strikingly different from those of the other five individuals, who had (relative) macrocephaly and hypertelorism. We measured the effect of SPOP variants on BET protein amounts in human Ishikawa endometrial cancer cells and patient-derived cell lines because we hypothesized that variants would lead to functional divergent effects on BET proteins. The de novo variants c.362G>A (p.Arg121Gln) and c. 430G>A (p.Asp144Asn), identified in the first two individuals, resulted in a gain of function, and conversely, the c.73A>G (p.Thr25Ala), c.248A>G (p.Tyr83Cys), c.395G>T (p.Gly132Val), and c.412C>T (p.Arg138Cys) variants resulted in a dominant-negative effect. Our findings suggest that these opposite functional effects caused by the variants in SPOP result in two distinct and clinically recognizable syndromic forms of intellectual disability with contrasting craniofacial dysmorphisms.


Asunto(s)
Mutación Missense , Trastornos del Neurodesarrollo/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Adolescente , Niño , Preescolar , Facies , Femenino , Humanos , Lactante , Discapacidad Intelectual/genética , Masculino , Cráneo/anomalías , Adulto Joven
3.
Cancer Lett ; 529: 11-18, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34974131

RESUMEN

Comprehensive cancer genome studies have revealed genetically-defined subtypes of prostate cancer with distinct truncal driver mutations. Because prostate cancer has been largely seen as a rather uniform disease, the clinical significance of this discovery remained largely obscure. However, recent findings imply distinct biological features and therapeutic vulnerabilities linked to specific truncal mutations. Here we review our current understanding of prostate cancers harboring recurrent point mutations in the ubiquitin ligase adaptor protein SPOP and discuss opportunities for future clinical translation. More specifically, activation of the androgen receptor (AR) signaling emerges as the key oncogenic pathway. SPOP-mutant prostate cancer patients respond to AR inhibition in various clinical settings. Molecular insights on how mutant SPOP promotes tumorigenesis may open more specific therapeutic avenues which, in combination with conventional AR-targeting agents, could improve the outcome of patients with SPOP-mutant prostate cancer.


Asunto(s)
Biomarcadores de Tumor , Mutación , Proteínas Nucleares/genética , Neoplasias de la Próstata/etiología , Proteínas Represoras/genética , Alelos , Toma de Decisiones Clínicas , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Mutación con Pérdida de Función , Masculino , Modelos Biológicos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Atención al Paciente , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Conformación Proteica , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transducción de Señal , Relación Estructura-Actividad
4.
Nat Commun ; 12(1): 734, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531470

RESUMEN

Driver genes with a mutually exclusive mutation pattern across tumor genomes are thought to have overlapping roles in tumorigenesis. In contrast, we show here that mutually exclusive prostate cancer driver alterations involving the ERG transcription factor and the ubiquitin ligase adaptor SPOP are synthetic sick. At the molecular level, the incompatible cancer pathways are driven by opposing functions in SPOP. ERG upregulates wild type SPOP to dampen androgen receptor (AR) signaling and sustain ERG activity through degradation of the bromodomain histone reader ZMYND11. Conversely, SPOP-mutant tumors stabilize ZMYND11 to repress ERG-function and enable oncogenic androgen receptor signaling. This dichotomy regulates the response to therapeutic interventions in the AR pathway. While mutant SPOP renders tumor cells susceptible to androgen deprivation therapies, ERG promotes sensitivity to high-dose androgen therapy and pharmacological inhibition of wild type SPOP. More generally, these results define a distinct class of antagonistic cancer drivers and a blueprint toward their therapeutic exploitation.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/metabolismo , Regulador Transcripcional ERG/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Mutación/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Neoplasias de la Próstata/genética , Unión Proteica , Proteómica , Receptores Androgénicos/metabolismo , Proteínas Represoras/genética , Transducción de Señal/fisiología , Regulador Transcripcional ERG/genética , Complejos de Ubiquitina-Proteína Ligasa/genética
5.
Nat Commun ; 12(1): 7033, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857732

RESUMEN

Comprehensive genomic studies have delineated key driver mutations linked to disease progression for most cancers. However, corresponding transcriptional changes remain largely elusive because of the bias associated with cross-study analysis. Here, we overcome these hurdles and generate a comprehensive prostate cancer transcriptome atlas that describes the roadmap to tumor progression in a qualitative and quantitative manner. Most cancers follow a uniform trajectory characterized by upregulation of polycomb-repressive-complex-2, G2-M checkpoints, and M2 macrophage polarization. Using patient-derived xenograft models, we functionally validate our observations and add single-cell resolution. Thereby, we show that tumor progression occurs through transcriptional adaption rather than a selection of pre-existing cancer cell clusters. Moreover, we determine at the single-cell level how inhibition of EZH2 - the top upregulated gene along the trajectory - reverts tumor progression and macrophage polarization. Finally, a user-friendly web-resource is provided enabling the investigation of dynamic transcriptional perturbations linked to disease progression.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/genética , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Transcriptoma , Animales , Atlas como Asunto , Línea Celular Tumoral , Progresión de la Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Proteínas de Neoplasias/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Análisis de Componente Principal , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Análisis de la Célula Individual
6.
Nat Med ; 23(9): 1046-1054, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28805821

RESUMEN

It is generally assumed that recurrent mutations within a given cancer driver gene elicit similar drug responses. Cancer genome studies have identified recurrent but divergent missense mutations affecting the substrate-recognition domain of the ubiquitin ligase adaptor SPOP in endometrial and prostate cancers. The therapeutic implications of these mutations remain incompletely understood. Here we analyzed changes in the ubiquitin landscape induced by endometrial cancer-associated SPOP mutations and identified BRD2, BRD3 and BRD4 proteins (BETs) as SPOP-CUL3 substrates that are preferentially degraded by endometrial cancer-associated SPOP mutants. The resulting reduction of BET protein levels sensitized cancer cells to BET inhibitors. Conversely, prostate cancer-specific SPOP mutations resulted in impaired degradation of BETs, promoting their resistance to pharmacologic inhibition. These results uncover an oncogenomics paradox, whereby mutations mapping to the same domain evoke opposing drug susceptibilities. Specifically, we provide a molecular rationale for the use of BET inhibitors to treat patients with endometrial but not prostate cancer who harbor SPOP mutations.


Asunto(s)
Adenocarcinoma de Células Claras/genética , Carcinoma Endometrioide/genética , Carcinosarcoma/genética , Neoplasias Endometriales/genética , Neoplasias Quísticas, Mucinosas y Serosas/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Acetanilidas/farmacología , Adenocarcinoma de Células Claras/metabolismo , Animales , Apoptosis/efectos de los fármacos , Azepinas/farmacología , Carcinoma Endometrioide/metabolismo , Carcinosarcoma/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Proteínas Cullin/metabolismo , Resistencia a Antineoplásicos , Neoplasias Endometriales/metabolismo , Epigénesis Genética , Femenino , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Masculino , Espectrometría de Masas , Ratones Desnudos , Terapia Molecular Dirigida , Mutación , Trasplante de Neoplasias , Neoplasias Quísticas, Mucinosas y Serosas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Ubiquitinación
7.
Pathol Res Pract ; 199(3): 165-70, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12812318

RESUMEN

Epithelioid hemangioendothelioma (EHE) is a rare low-grade malignant vascular neoplasm with unpredictable prognosis. We report on a patient suffering from a vascular neoplasm with primary manifestation in the skeleton and subsequent development of lesions of EHE in the spleen, liver and lung. Based on the assumption that involvement of visceral organs was due to metastatic spread, changes in clinical behavior, morphology, and proliferation index suggest malignant progression of the tumor. Analyzing the expression of genes known to be involved in the pathogenesis of vascular tumors, we found that the appearance of less differentiated tumor was paralleled by an accumulation of TP53 and murine double minute-2 (MDM-2) protein, decreased caveolin-1 (CAV-1) expression, and increased vascular endothelial growth factor (VEGF) expression. Mutations of the von-Hippel-Lindau-tumor-suppressor-gene (VHL) were excluded as mechanism of VEGF upregulation. Therefore, we propose that the expression of TP53, MDM-2, CAV-1 and VEGF as a marker of biologic behavior be verified in a larger case study of EHE.


Asunto(s)
Neoplasias Óseas/genética , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica , Hemangioendotelioma Epitelioide/genética , Hemangioendotelioma Epitelioide/patología , Proteínas Nucleares , Proteínas de Arabidopsis/fisiología , Biomarcadores de Tumor , Enfermedad Crónica , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Hemangioendotelioma Epitelioide/secundario , Humanos , Inmunohistoquímica , Hibridación in Situ , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Persona de Mediana Edad , Polimorfismo Conformacional Retorcido-Simple , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-mdm2 , Neoplasias del Bazo/secundario , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
8.
Science ; 346(6205): 85-89, 2014 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-25278611

RESUMEN

Cancer genome characterization has revealed driver mutations in genes that govern ubiquitylation; however, the mechanisms by which these alterations promote tumorigenesis remain incompletely characterized. Here, we analyzed changes in the ubiquitin landscape induced by prostate cancer-associated mutations of SPOP, an E3 ubiquitin ligase substrate-binding protein. SPOP mutants impaired ubiquitylation of a subset of proteins in a dominant-negative fashion. Of these, DEK and TRIM24 emerged as effector substrates consistently up-regulated by SPOP mutants. We highlight DEK as a SPOP substrate that exhibited decreases in ubiquitylation and proteasomal degradation resulting from heteromeric complexes of wild-type and mutant SPOP protein. DEK stabilization promoted prostate epithelial cell invasion, which implicated DEK as an oncogenic effector. More generally, these results provide a framework to decipher tumorigenic mechanisms linked to dysregulated ubiquitylation.


Asunto(s)
Proteínas Nucleares/genética , Neoplasias de la Próstata/metabolismo , Proteínas Represoras/genética , Ubiquitinación/genética , Secuencia de Bases , Sitios de Unión/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA