Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Biopolymers ; 115(3): e23576, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38511874

RESUMEN

EWSR1 (Ewing Sarcoma Related protein 1) is an RNA binding protein that is ubiquitously expressed across cell lines and involved in multiple parts of RNA processing, such as transcription, splicing, and mRNA transport. EWSR1 has also been implicated in cellular mechanisms to control formation of R-loops, a three-stranded nucleic acid structure consisting of a DNA:RNA hybrid and a displaced single-stranded DNA strand. Unscheduled R-loops result in genomic and transcription stress. Loss of function of EWSR1 functions commonly found in Ewing Sarcoma correlates with high abundance of R-loops. In this study, we investigated the mechanism for EWSR1 to recognize an R-loop structure specifically. Using electrophoretic mobility shift assays (EMSA), we detected the high affinity binding of EWSR1 to substrates representing components found in R-loops. EWSR1 specificity could be isolated to the DNA fork region, which transitions between double- and single-stranded DNA. Our data suggests that the Zinc-finger domain (ZnF) with flanking arginine and glycine rich (RGG) domains provide high affinity binding, while the RNA recognition motif (RRM) with its RGG domains offer improved specificity. This model offers a rational for EWSR1 specificity to encompass a wide range in contexts due to the DNA forks always found with R-loops.


Asunto(s)
ADN , Estructuras R-Loop , Proteína EWS de Unión a ARN , Proteína EWS de Unión a ARN/metabolismo , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/genética , Humanos , ADN/metabolismo , ADN/química , Unión Proteica , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/genética , Dedos de Zinc , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Ensayo de Cambio de Movilidad Electroforética
2.
Biopolymers ; 114(5): e23536, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36929870

RESUMEN

EWS is a member of the FET family of RNA/DNA binding proteins that regulate crucial phases of nucleic acid metabolism. EWS comprises an N-terminal low-complexity domain (LCD) and a C-terminal RNA-binding domain (RBD). The RBD is further divided into three RG-rich regions, which flank an RNA-recognition motif (RRM) and a zinc finger (ZnF) domain. Recently, EWS was shown to regulate R-loops in Ewing sarcoma, a pediatric bone and soft-tissue cancer in which a chromosomal translocation fuses the N-terminal LCD of EWS to the C-terminal DNA binding domain of the transcription factor FLI1. Though EWS was shown to directly bind R-loops, the binding mechanism was not elucidated. In the current study, the RBD of EWS was divided into several constructs, which were subsequently assayed for binding to various nucleic acid structures expected to form at R-loops, including RNA stem-loops, DNA G-quadruplexes, and RNA:DNA hybrids. EWS interacted with all three nucleic acid structures with varying affinities and multiple domains contributed to binding each substrate. The RRM and RG2 region appear to bind nucleic acids promiscuously while the ZnF displayed more selectivity for single-stranded structures. With these results, the structural underpinnings of EWS recognition and binding of R-loops and other nucleic acid structures is better understood.


Asunto(s)
Ácidos Nucleicos , Proteínas de Unión al ARN , Humanos , Niño , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Proteínas de Unión al ADN , ADN , ARN , Línea Celular Tumoral
3.
J Am Chem Soc ; 143(30): 11520-11534, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34304571

RESUMEN

Many membraneless organelles are thought to be biomolecular condensates formed by phase separation of proteins and other biopolymers. Post-translational modifications (PTMs) can impact protein phase separation behavior, although for many PTMs this aspect of their function is unknown. O-linked ß-D-N-acetylglucosaminylation (O-GlcNAcylation) is an abundant form of intracellular glycosylation whose roles in regulating biomolecular condensate assembly and dynamics have not been delineated. Using an in vitro approach, we found that O-GlcNAcylation reduces the phase separation propensity of the EWS N-terminal low complexity region (LCRN) under different conditions, including in the presence of the arginine- and glycine-rich RNA-binding domains (RBD). O-GlcNAcylation enhances fluorescence recovery after photobleaching (FRAP) within EWS LCRN condensates and causes the droplets to exhibit more liquid-like relaxation following fusion. Following extended incubation times, EWS LCRN+RBD condensates exhibit diminished FRAP, indicating a loss of fluidity, while condensates containing the O-GlcNAcylated LCRN do not. In HeLa cells, EWS is less O-GlcNAcylated following OGT knockdown, which correlates with its increased accumulation in a filter retardation assay. Relative to the human proteome, O-GlcNAcylated proteins are enriched with regions that are predicted to phase separate, suggesting a general role of O-GlcNAcylation in regulation of biomolecular condensates.


Asunto(s)
Acetilglucosamina/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Acetilglucosamina/química , Condensados Biomoleculares , Células HeLa , Humanos , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Proteína EWS de Unión a ARN/química , Células Tumorales Cultivadas
4.
Cancer Cell ; 39(9): 1262-1278.e7, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34329586

RESUMEN

Fusion-transcription factors (fusion-TFs) represent a class of driver oncoproteins that are difficult to therapeutically target. Recently, protein degradation has emerged as a strategy to target these challenging oncoproteins. The mechanisms that regulate fusion-TF stability, however, are generally unknown. Using CRISPR-Cas9 screening, we discovered tripartite motif-containing 8 (TRIM8) as an E3 ubiquitin ligase that ubiquitinates and degrades EWS/FLI, a driver fusion-TF in Ewing sarcoma. Moreover, we identified TRIM8 as a selective dependency in Ewing sarcoma compared with >700 other cancer cell lines. Mechanistically, TRIM8 knockout led to an increase in EWS/FLI protein levels that was not tolerated. EWS/FLI acts as a neomorphic substrate for TRIM8, defining the selective nature of the dependency. Our results demonstrate that fusion-TF protein stability is tightly regulated and highlight fusion oncoprotein-specific regulators as selective therapeutic targets. This study provides a tractable strategy to therapeutically exploit oncogene overdose in Ewing sarcoma and potentially other fusion-TF-driven cancers.


Asunto(s)
Neoplasias Óseas/mortalidad , Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Fusión Oncogénica/química , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/mortalidad , Neoplasias Óseas/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Fusión Oncogénica/metabolismo , Estabilidad Proteica , Proteolisis , Sarcoma de Ewing/metabolismo , Transactivadores/metabolismo
5.
Front Endocrinol (Lausanne) ; 12: 600682, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33692755

RESUMEN

Familial inheritance in non-medullary thyroid cancer (NMTC) is an area that has yet to be adequately explored. Despite evidence suggesting strong familial clustering of non-syndromic NMTC, known variants still account for a very small percentage of the genetic burden. In a recent whole genome sequencing (WGS) study of five families with several NMTCs, we shortlisted promising variants with the help of our in-house developed Familial Cancer Variant Prioritization Pipeline (FCVPPv2). Here, we report potentially disease-causing variants in checkpoint kinase 2 (CHEK2), Ewing sarcoma breakpoint region 1 (EWSR1) and T-lymphoma invasion and metastasis-inducing protein 1 (TIAM1) in one family. Performing WGS on three cases, one probable case and one healthy individual in a family with familial NMTC left us with 112254 variants with a minor allele frequency of less than 0.1%, which was reduced by pedigree-based filtering to 6368. Application of the pipeline led to the prioritization of seven coding and nine non-coding variants from this family. The variant identified in CHEK2, a known tumor suppressor gene involved in DNA damage-induced DNA repair, cell cycle arrest, and apoptosis, has been previously identified as a germline variant in breast and prostate cancer and has been functionally validated by Roeb et al. in a yeast-based assay to have an intermediate effect on protein function. We thus hypothesized that this family may harbor additional disease-causing variants in other functionally related genes. We evaluated two further variants in EWSR1 and TIAM1 with promising in silico results and reported interaction in the DNA-damage repair pathway. Hence, we propose a polygenic mode of inheritance in this family. As familial NMTC is considered to be more aggressive than its sporadic counterpart, it is important to identify such susceptibility genes and their associated pathways. In this way, the advancement of personalized medicine in NMTC patients can be fostered. We also wish to reopen the discussion on monogenic vs polygenic inheritance in NMTC and instigate further development in this area of research.


Asunto(s)
Quinasa de Punto de Control 2/genética , Predisposición Genética a la Enfermedad , Proteína EWS de Unión a ARN/genética , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/genética , Cáncer Papilar Tiroideo/genética , Secuencia de Aminoácidos , Quinasa de Punto de Control 2/química , Quinasa de Punto de Control 2/metabolismo , Femenino , Frecuencia de los Genes , Genoma Humano , Humanos , Italia , Masculino , Linaje , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Alineación de Secuencia , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/química , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T/metabolismo , Cáncer Papilar Tiroideo/metabolismo , Secuenciación Completa del Genoma
6.
J Vis Exp ; (160)2020 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-32658189

RESUMEN

Many cancers are characterized by chromosomal translocations which result in the expression of oncogenic fusion transcription factors. Typically, these proteins contain an intrinsically disordered domain (IDD) fused with the DNA-binding domain (DBD) of another protein and orchestrate widespread transcriptional changes to promote malignancy. These fusions are often the sole recurring genomic aberration in the cancers they cause, making them attractive therapeutic targets. However, targeting oncogenic transcription factors requires a better understanding of the mechanistic role that low-complexity, IDDs play in their function. The N-terminal domain of EWSR1 is an IDD involved in a variety of oncogenic fusion transcription factors, including EWS/FLI, EWS/ATF, and EWS/WT1. Here, we use RNA-sequencing to investigate the structural features of the EWS domain important for transcriptional function of EWS/FLI in Ewing sarcoma. First shRNA-mediated depletion of the endogenous fusion from Ewing sarcoma cells paired with ectopic expression of a variety of EWS-mutant constructs is performed. Then RNA-sequencing is used to analyze the transcriptomes of cells expressing these constructs to characterize the functional deficits associated with mutations in the EWS domain. By integrating the transcriptomic analyses with previously published information about EWS/FLI DNA binding motifs, and genomic localization, as well as functional assays for transforming ability, we were able to identify structural features of EWS/FLI important for oncogenesis and define a novel set of EWS/FLI target genes critical for Ewing sarcoma. This paper demonstrates the use of RNA-sequencing as a method to map the structure-function relationship of the intrinsically disordered domain of oncogenic transcription factors.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/química , Proteína EWS de Unión a ARN/metabolismo , Relación Estructura-Actividad , Sitios de Unión , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Humanos , Mutación , Proteínas de Fusión Oncogénica/genética , Dominios Proteicos , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología
7.
Nucleic Acids Res ; 47(18): 9619-9636, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31392992

RESUMEN

Connections between epigenetic reprogramming and transcription or splicing create novel mechanistic networks that can be targeted with tailored therapies. Multiple subunits of the chromatin remodeling BAF complex, including ARID1A, play a role in oncogenesis, either as tumor suppressors or oncogenes. Recent work demonstrated that EWS-FLI1, the oncogenic driver of Ewing sarcoma (ES), plays a role in chromatin regulation through interactions with the BAF complex. However, the specific BAF subunits that interact with EWS-FLI1 and the precise role of the BAF complex in ES oncogenesis remain unknown. In addition to regulating transcription, EWS-FLI1 also alters the splicing of many mRNA isoforms, but the role of splicing modulation in ES oncogenesis is not well understood. We have identified a direct connection between the EWS-FLI1 protein and ARID1A isoform protein variant ARID1A-L. We demonstrate here that ARID1A-L is critical for ES maintenance and supports oncogenic transformation. We further report a novel feed-forward cycle in which EWS-FLI1 leads to preferential splicing of ARID1A-L, promoting ES growth, and ARID1A-L reciprocally promotes EWS-FLI1 protein stability. Dissecting this interaction may lead to improved cancer-specific drug targeting.


Asunto(s)
Carcinogénesis/genética , Proteínas Nucleares/genética , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Factores de Transcripción/genética , Empalme Alternativo/genética , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Nucleares/química , Proteínas de Fusión Oncogénica/química , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estabilidad Proteica , Proteína Proto-Oncogénica c-fli-1/química , Proteína EWS de Unión a ARN/química , Sarcoma de Ewing/patología , Factores de Transcripción/química
8.
Sci Rep ; 9(1): 951, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700749

RESUMEN

Ewing sarcoma is the second most common pediatric bone and soft tissue tumor presenting with an aggressive behavior and prevalence to metastasize. The diagnostic translocation t(22;11)(q24;12) leads to expression of the chimeric oncoprotein EWS-FLI1 which is uniquely expressed in all tumor cells and maintains their survival. Constant EWS-FLI1 protein turnover is regulated by the ubiquitin proteasome system. Here, we now identified ubiquitin specific protease 19 (USP19) as a regulator of EWS-FLI1 stability using an siRNA based screening approach. Depletion of USP19 resulted in diminished EWS-FLI1 protein levels and, vice versa, upregulation of active USP19 stabilized the fusion protein. Importantly, stabilization appears to be specific for the fusion protein as it could not be observed neither for EWSR1 nor for FLI1 wild type proteins even though USP19 binds to the N-terminal EWS region to regulate deubiquitination of both EWS-FLI1 and EWSR1. Further, stable shUSP19 depletion resulted in decreased cell growth and diminished colony forming capacity in vitro, and significantly delayed tumor growth in vivo. Our findings not only provide novel insights into the importance of the N-terminal EWSR1 domain for regulation of fusion protein stability, but also indicate that inhibition of deubiquitinating enzyme(s) might constitute a novel therapeutic strategy in treatment of Ewing sarcoma.


Asunto(s)
Endopeptidasas/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Ubiquitinación , Animales , Proliferación Celular , Humanos , Ratones , Modelos Biológicos , Proteínas de Fusión Oncogénica/química , Dominios Proteicos , Estabilidad Proteica , Proteína Proto-Oncogénica c-fli-1/química , ARN Interferente Pequeño/metabolismo , Proteína EWS de Unión a ARN/química
9.
Cell ; 173(3): 677-692.e20, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677512

RESUMEN

RNA-binding proteins (RBPs) with prion-like domains (PrLDs) phase transition to functional liquids, which can mature into aberrant hydrogels composed of pathological fibrils that underpin fatal neurodegenerative disorders. Several nuclear RBPs with PrLDs, including TDP-43, FUS, hnRNPA1, and hnRNPA2, mislocalize to cytoplasmic inclusions in neurodegenerative disorders, and mutations in their PrLDs can accelerate fibrillization and cause disease. Here, we establish that nuclear-import receptors (NIRs) specifically chaperone and potently disaggregate wild-type and disease-linked RBPs bearing a NLS. Karyopherin-ß2 (also called Transportin-1) engages PY-NLSs to inhibit and reverse FUS, TAF15, EWSR1, hnRNPA1, and hnRNPA2 fibrillization, whereas Importin-α plus Karyopherin-ß1 prevent and reverse TDP-43 fibrillization. Remarkably, Karyopherin-ß2 dissolves phase-separated liquids and aberrant fibrillar hydrogels formed by FUS and hnRNPA1. In vivo, Karyopherin-ß2 prevents RBPs with PY-NLSs accumulating in stress granules, restores nuclear RBP localization and function, and rescues degeneration caused by disease-linked FUS and hnRNPA2. Thus, NIRs therapeutically restore RBP homeostasis and mitigate neurodegeneration.


Asunto(s)
Transporte Activo de Núcleo Celular , Priones/química , Proteínas de Unión al ARN/química , Receptores Citoplasmáticos y Nucleares/química , Adulto , Anciano , Animales , Citoplasma/química , Proteínas de Unión al ADN/química , Drosophila melanogaster , Femenino , Proteínas Fluorescentes Verdes/química , Células HEK293 , Células HeLa , Homeostasis , Humanos , Carioferinas/química , Masculino , Persona de Mediana Edad , Chaperonas Moleculares/química , Mutación , Enfermedades Neurodegenerativas/patología , Dominios Proteicos , Proteína EWS de Unión a ARN/química , Factores Asociados con la Proteína de Unión a TATA/química , beta Carioferinas/química
10.
Protein Sci ; 27(3): 633-642, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29193371

RESUMEN

The FET sub-family (FUS/TLS, EWS, TAF15) of RNA-binding proteins have remarkably similar overall structure but diverse biological and pathological roles. The molecular basis for FET protein specialization is largely unknown. Gly-Arg-Rich regions (RGG-boxes) within FET proteins are targets for methylation by Protein-Arginine-Methyl-Transferase-1 (PRMT1) and substrate capture is thought to involve electrostatic attraction between positively charged polyRGG substrates and negatively charged surface channels of PRMT1. Unlike FUS and EWS, a high proportion of TAF15 RGG-boxes are embedded within neutrally charged YGGDR(S/G)G repeats, suggesting that they might not bind well to PRMT1. This notion runs contrary however to a report that YGGDR(S/G)G repeats are methylated by PRMT1. Using peptide-based polyRGG substrates and a novel 2-hybrid binding assay, we find that the Asp residue in YGGDR(S/G)G repeats confers poor binding to PRMT1. Our results therefore indicate that YGGDR(S/G)G repeats may contribute to TAF15 specialization by enabling differential interactions with PRMT1 and reduced overall levels of TAF15 methylation compared with other FET proteins. By analogy with molecular recognition of other disordered polyvalent ligands by globular protein partners, we also propose a dynamic polyelectrostatic model for substrate capture by PRMT1.


Asunto(s)
Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Proteínas Represoras/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Asparagina/metabolismo , Sitios de Unión , Línea Celular , Humanos , Metilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína EWS de Unión a ARN/química , Factores Asociados con la Proteína de Unión a TATA/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA