Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EMBO J ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951610

RESUMEN

Cells have evolved a robust and highly regulated DNA damage response to preserve their genomic integrity. Although increasing evidence highlights the relevance of RNA regulation, our understanding of its impact on a fully efficient DNA damage response remains limited. Here, through a targeted CRISPR-knockout screen, we identify RNA-binding proteins and modifiers that participate in the p53 response. Among the top hits, we find the m6A reader YTHDC1 as a master regulator of p53 expression. YTHDC1 binds to the transcription start sites of TP53 and other genes involved in the DNA damage response, promoting their transcriptional elongation. YTHDC1 deficiency also causes the retention of introns and therefore aberrant protein production of key DNA damage factors. While YTHDC1-mediated intron retention requires m6A, TP53 transcriptional pause-release is promoted by YTHDC1 independently of m6A. Depletion of YTHDC1 causes genomic instability and aberrant cancer cell proliferation mediated by genes regulated by YTHDC1. Our results uncover YTHDC1 as an orchestrator of the DNA damage response through distinct mechanisms of co-transcriptional mRNA regulation.

2.
Mol Cell ; 63(3): 397-407, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27477908

RESUMEN

Long noncoding RNAs (lncRNAs) are involved in diverse cellular processes through multiple mechanisms. Here, we describe a previously uncharacterized human lncRNA, CONCR (cohesion regulator noncoding RNA), that is transcriptionally activated by MYC and is upregulated in multiple cancer types. The expression of CONCR is cell cycle regulated, and it is required for cell-cycle progression and DNA replication. Moreover, cells depleted of CONCR show severe defects in sister chromatid cohesion, suggesting an essential role for CONCR in cohesion establishment during cell division. CONCR interacts with and regulates the activity of DDX11, a DNA-dependent ATPase and helicase involved in DNA replication and sister chromatid cohesion. These findings unveil a direct role for an lncRNA in the establishment of sister chromatid cohesion by modulating DDX11 enzymatic activity.


Asunto(s)
Cromátides/metabolismo , Replicación del ADN , ADN de Neoplasias/biosíntesis , Neoplasias/metabolismo , ARN Largo no Codificante/metabolismo , Células A549 , Animales , Apoptosis , Proliferación Celular , Cromátides/genética , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN de Neoplasias/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Ratones Endogámicos BALB C , Ratones Transgénicos , Neoplasias/genética , Neoplasias/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , ARN Largo no Codificante/genética , Factores de Tiempo , Transcripción Genética , Activación Transcripcional , Transfección , Carga Tumoral , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Biochem Biophys Res Commun ; 483(1): 516-521, 2017 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-28007599

RESUMEN

The TP53 gene is the most frequently mutated gene in human cancer and p53 protein plays a crucial role in gene expression and cancer protection. Its role is manifested by interactions with other proteins and DNA. p53 is a transcription factor that binds to DNA response elements (REs). Due to the palindromic nature of the consensus binding site, several p53-REs have the potential to form cruciform structures. However, the influence of cruciform formation on the activity of p53-REs has not been evaluated. Therefore, we prepared sets of p53-REs with identical theoretical binding affinity in their linear state, but different probabilities to form extra helical structures, for in vitro and in vivo analyses. Then we evaluated the presence of cruciform structures when inserted into plasmid DNA and employed a yeast-based assay to measure transactivation potential of these p53-REs cloned at a chromosomal locus in isogenic strains. We show that transactivation in vivo correlated more with relative propensity of an RE to form cruciforms than to its predicted in vitro DNA binding affinity for wild type p53. Structural features of p53-REs could therefore be an important determinant of p53 transactivation function.


Asunto(s)
Secuencias Invertidas Repetidas , Elementos de Respuesta , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Cromatina/genética , Simulación por Computador , Mutación , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Levaduras/genética
4.
EMBO Mol Med ; 16(1): 112-131, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38182795

RESUMEN

The therapeutic use of adeno-associated viral vector (AAV)-mediated gene disruption using CRISPR-Cas9 is limited by potential off-target modifications and the risk of uncontrolled integration of vector genomes into CRISPR-mediated double-strand breaks. To address these concerns, we explored the use of AAV-delivered paired Staphylococcus aureus nickases (D10ASaCas9) to target the Hao1 gene for the treatment of primary hyperoxaluria type 1 (PH1). Our study demonstrated effective Hao1 gene disruption, a significant decrease in glycolate oxidase expression, and a therapeutic effect in PH1 mice. The assessment of undesired genetic modifications through CIRCLE-seq and CAST-Seq analyses revealed neither off-target activity nor chromosomal translocations. Importantly, the use of paired-D10ASaCas9 resulted in a significant reduction in AAV integration at the target site compared to SaCas9 nuclease. In addition, our study highlights the limitations of current analytical tools in characterizing modifications introduced by paired D10ASaCas9, necessitating the development of a custom pipeline for more accurate characterization. These results describe a positive advance towards a safe and effective potential long-term treatment for PH1 patients.


Asunto(s)
Sistemas CRISPR-Cas , Hiperoxaluria Primaria , Humanos , Animales , Ratones , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Edición Génica , Hiperoxaluria Primaria/genética , Hiperoxaluria Primaria/terapia
5.
Nat Biotechnol ; 41(6): 806-812, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36536150

RESUMEN

Chromatin states are functionally defined by a complex combination of histone modifications, transcription factor binding, DNA accessibility and other factors. Current methods for defining chromatin states cannot measure more than one aspect in a single experiment at single-cell resolution. Here we introduce nanobody-tethered transposition followed by sequencing (NTT-seq), an assay capable of measuring the genome-wide presence of up to three histone modifications and protein-DNA binding sites at single-cell resolution. NTT-seq uses recombinant Tn5 transposase fused to a set of secondary nanobodies (nb). Each nb-Tn5 fusion protein specifically binds to different immunoglobulin-G antibodies, enabling a mixture of primary antibodies binding different epitopes to be used in a single experiment. We apply bulk-cell and single-cell NTT-seq to generate high-resolution multimodal maps of chromatin states in cell culture and in human immune cells. We also extend NTT-seq to enable simultaneous profiling of cell surface protein expression and multimodal chromatin states to study cells of the immune system.


Asunto(s)
Cromatina , ADN , Humanos , Cromatina/genética , ADN/metabolismo , Análisis de Secuencia de ADN/métodos , Genoma , Unión Proteica , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de la Célula Individual
6.
Nat Biotechnol ; 40(8): 1220-1230, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35332340

RESUMEN

Technologies that profile chromatin modifications at single-cell resolution offer enormous promise for functional genomic characterization, but the sparsity of the measurements and integrating multiple binding maps represent substantial challenges. Here we introduce single-cell (sc)CUT&Tag-pro, a multimodal assay for profiling protein-DNA interactions coupled with the abundance of surface proteins in single cells. In addition, we introduce single-cell ChromHMM, which integrates data from multiple experiments to infer and annotate chromatin states based on combinatorial histone modification patterns. We apply these tools to perform an integrated analysis across nine different molecular modalities in circulating human immune cells. We demonstrate how these two approaches can characterize dynamic changes in the function of individual genomic elements across both discrete cell states and continuous developmental trajectories, nominate associated motifs and regulators that establish chromatin states and identify extensive and cell-type-specific regulatory priming. Finally, we demonstrate how our integrated reference can serve as a scaffold to map and improve the interpretation of additional scCUT&Tag datasets.


Asunto(s)
Cromatina , Histonas , Cromatina/genética , Inmunoprecipitación de Cromatina , ADN , Genómica , Histonas/genética , Histonas/metabolismo , Humanos
7.
Biotechnol Appl Biochem ; 58(1): 39-49, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21446958

RESUMEN

We report the expression of recombinant RNASET2, the only human member of the Rh/T2/S family of acid ribonucleases, in the yeast Pichia pastoris and the baculovirus-insect cell heterologous systems. In both models, the yield of recombinant protein was comparable and ranged between 5 mg/L (for a catalytically impaired mutant version of RNASET2) and 30 mg/L for the wild-type protein. Thus, the produced protein version rather than the expression system used appears to influence protein yield after optimization of culture conditions. The recombinant protein was found to undergo heterogeneous glycosylation in both systems, particularly in P. pastoris. Most importantly, the wild-type protein purified from both systems was found to be catalytically competent. The expression of recombinant RNASET2 in both systems will allow the implementation of functional assays in vivo and in vitro to better define the antioncogenic properties of this member of the Rh/T2/S ribonuclease family.


Asunto(s)
Baculoviridae/metabolismo , Regulación Neoplásica de la Expresión Génica , Pichia/metabolismo , Ribonucleasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Baculoviridae/genética , Secuencia de Bases , Biocatálisis , Células Cultivadas , Clonación Molecular , Glicosilación , Humanos , Datos de Secuencia Molecular , Mutación , Pichia/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ribonucleasas/genética , Proteínas Supresoras de Tumor/genética
8.
Cancer Res ; 81(6): 1457-1471, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33372039

RESUMEN

Neuroblastoma has a low mutation rate for the p53 gene. Alternative ways of p53 inactivation have been proposed in neuroblastoma, such as abnormal cytoplasmic accumulation of wild-type p53. However, mechanisms leading to p53 inactivation via cytoplasmic accumulation are not well investigated. Here we show that the neuroblastoma risk-associated locus 6p22.3-derived tumor suppressor NBAT1 is a p53-responsive lncRNA that regulates p53 subcellular levels. Low expression of NBAT1 provided resistance to genotoxic drugs by promoting p53 accumulation in cytoplasm and loss from mitochondrial and nuclear compartments. Depletion of NBAT1 altered CRM1 function and contributed to the loss of p53-dependent nuclear gene expression during genotoxic drug treatment. CRM1 inhibition rescued p53-dependent nuclear functions and sensitized NBAT1-depleted cells to genotoxic drugs. Combined inhibition of CRM1 and MDM2 was even more effective in sensitizing aggressive neuroblastoma cells with p53 cytoplasmic accumulation. Thus, our mechanistic studies uncover an NBAT1-dependent CRM1/MDM2-based potential combination therapy for patients with high-risk neuroblastoma. SIGNIFICANCE: This study shows how a p53-responsive lncRNA mediates chemotherapeutic response by modulating nuclear p53 pathways and identifies a potential treatment strategy for patients with high-risk neuroblastoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/genética , Neuroblastoma/tratamiento farmacológico , ARN Largo no Codificante/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis , Fraccionamiento Celular , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Daño del ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Masculino , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Neuroblastoma/genética , Neuroblastoma/patología , Neuroblastoma/cirugía , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Largo no Codificante/genética , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
9.
Nat Commun ; 11(1): 936, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071317

RESUMEN

LncRNAs have been shown to be direct players in chromatin regulation, but little is known about their role at active genomic loci. We investigate the role of lncRNAs in gene activation by profiling the RNA interactome of SMARCB1-containing SWI/SNF complexes in proliferating and senescent conditions. The isolation of SMARCB1-associated transcripts, together with chromatin profiling, shows prevalent association to active regions where SMARCB1 differentially binds locally transcribed RNAs. We identify SWINGN, a lncRNA interacting with SMARCB1 exclusively in proliferating conditions, exerting a pro-oncogenic role in some tumor types. SWINGN is transcribed from an enhancer and modulates the activation of GAS6 oncogene as part of a topologically organized region, as well as a larger network of pro-oncogenic genes by favoring SMARCB1 binding. Our results indicate that SWINGN influences the ability of the SWI/SNF complexes to drive epigenetic activation of specific promoters, suggesting a SWI/SNF-RNA cooperation to achieve optimal transcriptional activation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Regiones Promotoras Genéticas/genética , ARN Largo no Codificante/metabolismo , Proteína SMARCB1/metabolismo , Animales , Apoptosis/genética , Carcinogénesis , Proliferación Celular/genética , Conjuntos de Datos como Asunto , Femenino , Redes Reguladoras de Genes , Células HCT116 , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Neoplasias/patología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , RNA-Seq , Activación Transcripcional , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Cell Biol ; 219(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32858747

RESUMEN

Cancer is characterized by genomic instability leading to deletion or amplification of oncogenes or tumor suppressors. However, most of the altered regions are devoid of known cancer drivers. Here, we identify lncRNAs frequently lost or amplified in cancer. Among them, we found amplified lncRNA associated with lung cancer-1 (ALAL-1) as frequently amplified in lung adenocarcinomas. ALAL-1 is also overexpressed in additional tumor types, such as lung squamous carcinoma. The RNA product of ALAL-1 is able to promote the proliferation and tumorigenicity of lung cancer cells. ALAL-1 is a TNFα- and NF-κB-induced cytoplasmic lncRNA that specifically interacts with SART3, regulating the subcellular localization of the protein deubiquitinase USP4 and, in turn, its function in the cell. Interestingly, ALAL-1 expression inversely correlates with the immune infiltration of lung squamous tumors, while tumors with ALAL-1 amplification show lower infiltration of several types of immune cells. We have thus unveiled a pro-oncogenic lncRNA that mediates cancer immune evasion, pointing to a new target for immune potentiation.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Evasión Inmune/genética , Neoplasias Pulmonares/genética , ARN Largo no Codificante/genética , Células A549 , Adenocarcinoma del Pulmón/genética , Antígenos de Neoplasias/genética , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , FN-kappa B/genética , Oncogenes/genética , Proteasas Ubiquitina-Específicas/genética
11.
Genome Biol ; 18(1): 206, 2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29084573

RESUMEN

A major shift in our understanding of genome regulation has emerged recently. It is now apparent that the majority of cellular transcripts do not code for proteins, and many of them are long noncoding RNAs (lncRNAs). Increasingly, studies suggest that lncRNAs regulate gene expression through diverse mechanisms. We review emerging mechanistic views of lncRNAs in gene regulation in the cell nucleus. We discuss the functional interactions that lncRNAs establish with other molecules as well as the relationship between lncRNA transcription and function. While some of these mechanisms are specific to lncRNAs, others might be shared with other types of genes.


Asunto(s)
Regulación de la Expresión Génica , ARN Largo no Codificante/metabolismo , Animales , Cromatina/metabolismo , ADN/química , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Proteínas de Unión al ARN/metabolismo , Transcripción Genética
12.
Genome Biol ; 18(1): 202, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-29078818

RESUMEN

BACKGROUND: It is now obvious that the majority of cellular transcripts do not code for proteins, and a significant subset of them are long non-coding RNAs (lncRNAs). Many lncRNAs show aberrant expression in cancer, and some of them have been linked to cell transformation. However, the underlying mechanisms remain poorly understood and it is unknown how the sequences of lncRNA dictate their function. RESULTS: Here we characterize the function of the p53-regulated human lncRNA LINC-PINT in cancer. We find that LINC-PINT is downregulated in multiple types of cancer and acts as a tumor suppressor lncRNA by reducing the invasive phenotype of cancer cells. A cross-species analysis identifies a highly conserved sequence element in LINC-PINT that is essential for its function. This sequence mediates a specific interaction with PRC2, necessary for the LINC-PINT-dependent repression of a pro-invasion signature of genes regulated by the transcription factor EGR1. CONCLUSIONS: Our findings support a conserved functional co-dependence between LINC-PINT and PRC2 and lead us to propose a new mechanism where the lncRNA regulates the availability of free PRC2 at the proximity of co-regulated genomic loci.


Asunto(s)
Invasividad Neoplásica , ARN Largo no Codificante/química , ARN Largo no Codificante/fisiología , Animales , Secuencia de Bases , Movimiento Celular , Secuencia Conservada , Regulación hacia Abajo , Silenciador del Gen , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Complejo Represivo Polycomb 2/metabolismo
13.
Cancer Res ; 76(9): 2626-36, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197265

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is a core process underlying cell movement during embryonic development and morphogenesis. Cancer cells hijack this developmental program to execute a multi-step cascade, leading to tumorigenesis and metastasis. CD133 (PROM1), a marker of cancer stem cells, has been shown to facilitate EMT in various cancers, but the regulatory networks controlling CD133 gene expression and function in cancer remain incompletely delineated. In this study, we show that a ribonucleoprotein complex including the long noncoding RNA MALAT1 and the RNA-binding protein HuR (ELAVL1) binds the CD133 promoter region to regulate its expression. In luminal nonmetastatic MCF-7 breast cancer cells, HuR silencing was sufficient to upregulate N-cadherin (CDH2) and CD133 along with a migratory and mesenchymal-like phenotype. Furthermore, we found that in the basal-like metastatic cell line MDA-MB-231 and primary triple-negative breast cancer tumor cells, the repressor complex was absent from the CD133-regulatory region, but was present in the MCF-7 and primary ER+ tumor cells. The absence of the complex from basal-like cells was attributed to diminished expression of MALAT1, which, when overexpressed, dampened CD133 levels. In conclusion, our findings suggest that the failure of a repressive complex to form or stabilize in breast cancer promotes CD133 upregulation and an EMT-like program, providing new mechanistic insights underlying the control of prometastatic processes. Cancer Res; 76(9); 2626-36. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Proteína 1 Similar a ELAV/metabolismo , Transición Epitelial-Mesenquimal/fisiología , ARN Largo no Codificante/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
PLoS One ; 10(2): e0116177, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25668429

RESUMEN

Co-evolution of transcription factors (TFs) with their respective cis-regulatory network enhances functional diversity in the course of evolution. We present a new approach to investigate transactivation capacity of sequence-specific TFs in evolutionary studies. Saccharomyces cerevisiae was used as an in vivo test tube and p53 proteins derived from human and five commonly used animal models were chosen as proof of concept. p53 is a highly conserved master regulator of environmental stress responses. Previous reports indicated conserved p53 DNA binding specificity in vitro, even for evolutionary distant species. We used isogenic yeast strains where p53-dependent transactivation was measured towards chromosomally integrated p53 response elements (REs). Ten REs were chosen to sample a wide range of DNA binding affinity and transactivation capacity for human p53 and proteins were expressed at two levels using an inducible expression system. We showed that the assay is amenable to study thermo-sensitivity of frog p53, and that chimeric constructs containing an ectopic transactivation domain could be rapidly developed to enhance the activity of proteins, such as fruit fly p53, that are poorly effective in engaging the yeast transcriptional machinery. Changes in the profile of relative transactivation towards the ten REs were measured for each p53 protein and compared to the profile obtained with human p53. These results, which are largely independent from relative p53 protein levels, revealed widespread evolutionary divergence of p53 transactivation specificity, even between human and mouse p53. Fruit fly and human p53 exhibited the largest discrimination among REs while zebrafish p53 was the least selective.


Asunto(s)
Evolución Molecular , Elementos de Respuesta/genética , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Análisis por Conglomerados , Drosophila , Humanos , Isoquinolinas , Ratones , Datos de Secuencia Molecular , Filogenia , Plásmidos/genética , Saccharomyces cerevisiae , Alineación de Secuencia , Análisis de Secuencia de ADN , Especificidad de la Especie , Xenopus laevis , Pez Cebra
15.
Oncotarget ; 5(23): 12111-25, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25401416

RESUMEN

The p53 and NFκB sequence-specific transcription factors play crucial roles in cell proliferation and survival with critical, even if typically opposite, effects on cancer progression. To investigate a possible crosstalk between p53 and NFκB driven by chemotherapy-induced responses in the context of an inflammatory microenvironment, we performed a proof of concept study using MCF7 cells. Transcriptome analyses upon single or combined treatments with doxorubicin (Doxo, 1.5µM) and the NFκB inducer TNF-alpha (TNFα, 5ng/ml) revealed 432 up-regulated (log2 FC> 2), and 390 repressed genes (log2 FC< -2) for the Doxo+TNFα treatment. 239 up-regulated and 161 repressed genes were synergistically regulated by the double treatment. Annotation and pathway analyses of Doxo+TNFα selectively up-regulated genes indicated strong enrichment for cell migration terms. A panel of genes was examined by qPCR coupled to p53 activation by Doxo, 5-Fluoruracil and Nutlin-3a, or to p53 or NFκB inhibition. Transcriptome data were confirmed for 12 of 15 selected genes and seven (PLK3, LAMP3, ETV7, UNC5B, NTN1, DUSP5, SNAI1) were synergistically up-regulated after Doxo+TNFα and dependent both on p53 and NFκB. Migration assays consistently showed an increase in motility for MCF7 cells upon Doxo+TNFα. A signature of 29 Doxo+TNFα highly synergistic genes exhibited prognostic value for luminal breast cancer patients, with adverse outcome correlating with higher relative expression. We propose that the crosstalk between p53 and NFκB can lead to the activation of specific gene expression programs that may impact on cancer phenotypes and potentially modify the efficacy of cancer therapy.


Asunto(s)
Neoplasias de la Mama/patología , Expresión Génica/genética , FN-kappa B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Doxorrubicina/farmacología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma , Factor de Necrosis Tumoral alfa/farmacología
16.
Oncotarget ; 5(8): 2116-30, 2014 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-24926492

RESUMEN

TP63 is a member of the TP53 gene family that encodes for up to ten different TA and ∆N isoforms through alternative promoter usage and alternative splicing. Besides being a master regulator of gene expression for squamous epithelial proliferation, differentiation and maintenance, P63, through differential expression of its isoforms, plays important roles in tumorigenesis. All P63 isoforms share an immunoglobulin-like folded DNA binding domain responsible for binding to sequence-specific response elements (REs), whose overall consensus sequence is similar to that of the canonical p53 RE. Using a defined assay in yeast, where P63 isoforms and RE sequences are the only variables, and gene expression assays in human cell lines, we demonstrated that human TA- and ∆N-P63α proteins exhibited differences in transactivation specificity not observed with the corresponding P73 or P53 protein isoforms. These differences 1) were dependent on specific features of the RE sequence, 2) could be related to intrinsic differences in their oligomeric state and cooperative DNA binding, and 3) appeared to be conserved in evolution. Sicen genotoxic stress can change relative ratio of TA- and ∆N-P63α protein levels, the different transactivation specificity of each P63 isoform could potentially influence cellular responses to specific stresses.


Asunto(s)
ADN/química , ADN/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Proteínas Supresoras de Tumor/metabolismo , Western Blotting , Proteínas de Unión al ADN/metabolismo , Células HCT116 , Humanos , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas/metabolismo , Elementos de Respuesta/fisiología , Transfección
17.
PLoS One ; 8(7): e69152, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861960

RESUMEN

The tumor suppressor p53 was previously shown to markedly up-regulate the expression of the PRODH gene, encoding the proline dehydrogenase (PRODH) enzyme, which catalyzes the first step in proline degradation. Also PRODH2, which degrades 4-hydroxy-L-proline, a product of protein (e.g. collagen) catabolism, was recently described as a p53 target. Here, we confirmed p53-dependent induction of endogenous PRODH in response to genotoxic damage in cell lines of different histological origin. We established that over-expression of TAp73ß or TAp63ß is sufficient to induce PRODH expression in p53-null cells and that PRODH expression parallels the modulation of endogenous p73 by genotoxic drugs in several cell lines. The p53, p63, and p73-dependent transcriptional activation was linked to specific intronic response elements (REs), among those predicted by bioinformatics tools and experimentally validated by a yeast-based transactivation assay. p53 occupancy measurements were validated in HCT116 and MCF7 human cell lines. Conversely, PRODH2 was not responsive to p63 nor p73 and, at best, could be considered a weak p53 target. In fact, minimal levels of PRODH2 transcript induction by genotoxic stress was observed exclusively in one of four p53 wild-type cell lines tested. Consistently, all predicted p53 REs in PRODH2 were poor matches to the p53 RE consensus and showed very weak responsiveness, only to p53, in the functional assay. Taken together, our results highlight that PRODH, but not PRODH2, expression is under the control of p53 family members, specifically p53 and p73. This supports a deeper link between proteins of the p53-family and metabolic pathways, as PRODH modulates the balance of proline and glutamate levels and those of their derivative alpha-keto-glutarate (α-KG) under normal and pathological (tumor) conditions.


Asunto(s)
Intrones/genética , Prolina Oxidasa/metabolismo , Elementos de Respuesta/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Bases , Sitios de Unión/genética , Daño del ADN/genética , Células HCT116 , Humanos , Células MCF-7 , Datos de Secuencia Molecular , Prolina Oxidasa/genética , Unión Proteica/genética , Estabilidad Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA