Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Mol Cell ; 75(6): 1092-1101, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31539507

RESUMEN

Landmark discoveries made nearly two decades ago identified known transcriptional regulators as histone lysine methyltransferases. Since then, the field of lysine methylation signaling has been dominated by studies of how this small chemical posttranslational modification regulates gene expression and other chromatin-based processes. However, recent advances in mass-spectrometry-based proteomics have revealed that histones are just a subset of the thousands of eukaryotic proteins marked by lysine methylation. As the writers, erasers, and readers of histone lysine methylation are emerging as a promising therapeutic target class for cancer and other diseases, a key challenge for the field is to define the full spectrum of activities for these proteins. Here we summarize recent discoveries implicating non-histone lysine methylation as a major regulator of diverse cellular processes. We further discuss recent technological innovations that are enabling the expanded study of lysine methylation signaling. Collectively, these findings are shaping our understanding of the fundamental mechanisms of non-histone protein regulation through this dynamic and multi-functional posttranslational modification.


Asunto(s)
Epigenoma , Lisina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Animales , Humanos , Metilación
2.
Genome Res ; 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37918959

RESUMEN

Point mutations within the TERT promoter are the most recurrent somatic noncoding mutations identified across different cancer types, including glioblastoma, melanoma, hepatocellular carcinoma, and bladder cancer. They are most abundant at -146C > T and -124C > T, and rarer at -57A > C, with the latter originally described as a familial case, but subsequently shown also to occur somatically. All three mutations create de novo E26-specific (ETS) binding sites and result in activation of the TERT gene, allowing cancer cells to achieve replicative immortality. Here, we used a systematic proteomics screen to identify transcription factors preferentially binding to the -146C > T, -124C > T, and -57A > C mutations. Although we confirmed binding of multiple ETS factors to the mutant -146C > T and -124C > T sequences, we identified E4F1 as a -57A > C-specific binder and ZNF148 as a TERT wild-type (WT) promoter binder that showed reduced interaction with the -124C > T allele. Both proteins are activating transcription factors that bind specifically to the -57A > C and WT (at position 124) TERT promoter sequence in corresponding cell lines, and up-regulate TERT transcription and telomerase activity. Our work describes new regulators of TERT gene expression with possible roles in cancer.

3.
Mol Cell ; 67(4): 550-565.e5, 2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28803780

RESUMEN

DNA methylation is an essential epigenetic mark in mammals that has to be re-established after each round of DNA replication. The protein UHRF1 is essential for this process; it has been proposed that the protein targets newly replicated DNA by cooperatively binding hemi-methylated DNA and H3K9me2/3, but this model leaves a number of questions unanswered. Here, we present evidence for a direct recruitment of UHRF1 by the replication machinery via DNA ligase 1 (LIG1). A histone H3K9-like mimic within LIG1 is methylated by G9a and GLP and, compared with H3K9me2/3, more avidly binds UHRF1. Interaction with methylated LIG1 promotes the recruitment of UHRF1 to DNA replication sites and is required for DNA methylation maintenance. These results further elucidate the function of UHRF1, identify a non-histone target of G9a and GLP, and provide an example of a histone mimic that coordinates DNA replication and DNA methylation maintenance.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , ADN Ligasa (ATP)/metabolismo , Metilación de ADN , Replicación del ADN , ADN/biosíntesis , Epigénesis Genética , Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Proteínas Potenciadoras de Unión a CCAAT/química , Proteínas Potenciadoras de Unión a CCAAT/genética , ADN/genética , ADN Ligasa (ATP)/química , ADN Ligasa (ATP)/genética , Células Madre Embrionarias/enzimología , Células HEK293 , Células HeLa , Antígenos de Histocompatibilidad/química , Antígenos de Histocompatibilidad/genética , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Humanos , Lisina , Metilación , Ratones , Modelos Moleculares , Imitación Molecular , Mutación , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad , Transfección , Dominio Tudor , Ubiquitina-Proteína Ligasas
4.
Nucleic Acids Res ; 51(19): 10292-10308, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37650637

RESUMEN

Epigenetic mechanisms are essential to establish and safeguard cellular identities in mammals. They dynamically regulate the expression of genes, transposable elements and higher-order chromatin structures. Consequently, these chromatin marks are indispensable for mammalian development and alterations often lead to disease, such as cancer. Bivalent promoters are especially important during differentiation and development. Here we used a genetic screen to identify new regulators of a bivalent repressed gene. We identify BEND3 as a regulator of hundreds of bivalent promoters, some of which it represses, and some of which it activates. We show that BEND3 is recruited to a CpG-containg consensus site that is present in multiple copies in many bivalent promoters. Besides having direct effect on the promoters it binds, the loss of BEND3 leads to genome-wide gains of DNA methylation, which are especially marked at regions normally protected by the TET enzymes. DNA hydroxymethylation is reduced in Bend3 mutant cells, possibly as consequence of altered gene expression leading to diminished alpha-ketoglutarate production, thus lowering TET activity. Our results clarify the direct and indirect roles of an important chromatin regulator, BEND3, and, more broadly, they shed light on the regulation of bivalent promoters.


Asunto(s)
Metilación de ADN , Proteínas Represoras , Animales , Humanos , Cromatina/genética , Metilación de ADN/genética , Epigénesis Genética , Expresión Génica , Mamíferos/genética , Neoplasias/genética , Proteínas Represoras/metabolismo
5.
Nucleic Acids Res ; 49(6): 3020-3032, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33300031

RESUMEN

DNA methylation is essential to development and cellular physiology in mammals. Faulty DNA methylation is frequently observed in human diseases like cancer and neurological disorders. Molecularly, this epigenetic mark is linked to other chromatin modifications and it regulates key genomic processes, including transcription and splicing. Each round of DNA replication generates two hemi-methylated copies of the genome. These must be converted back to symmetrically methylated DNA before the next S-phase, or the mark will fade away; therefore the maintenance of DNA methylation is essential. Mechanistically, the maintenance of this epigenetic modification takes place during and after DNA replication, and occurs within the very dynamic context of chromatin re-assembly. Here, we review recent discoveries and unresolved questions regarding the mechanisms, dynamics and fidelity of DNA methylation maintenance in mammals. We also discuss how it could be regulated in normal development and misregulated in disease.


Asunto(s)
Metilación de ADN , Mamíferos/genética , Animales , Ensamble y Desensamble de Cromatina , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Replicación del ADN , Epigénesis Genética , Humanos , Neoplasias/genética , Enfermedades del Sistema Nervioso/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Bioorg Med Chem ; 52: 116500, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34801826

RESUMEN

The accumulation of epigenetic alterations is one of the major causes of tumorigenesis. Aberrant DNA methylation patterns cause genome instability and silencing of tumor suppressor genes in various types of tumors. Therefore, drugs that target DNA methylation-regulating factors have great potential for cancer therapy. Ubiquitin-like containing PHD and RING finger domain 1 (UHRF1) is an essential factor for DNA methylation maintenance. UHRF1 is overexpressed in various cancer cells and down-regulation of UHRF1 in these cells reactivates the expression of tumor suppressor genes, thus UHRF1 is a promising target for cancer therapy. We have previously shown that interaction between the tandem Tudor domain (TTD) of UHRF1 and DNA ligase 1 (LIG1) di/trimethylated on Lys126 plays a key role in the recruitment of UHRF1 to replication sites and replication-coupled DNA methylation maintenance. An arginine binding cavity (Arg-binding cavity) of the TTD is essential for LIG1 interaction, thus the development of inhibitors that target the Arg-binding cavity could potentially repress UHRF1 function in cancer cells. To develop such an inhibitor, we performed in silico screening using not only static but also dynamic metrics based on all-atom molecular dynamics simulations, resulting in efficient identification of 5-amino-2,4-dimethylpyridine (5A-DMP) as a novel TTD-binding compound. Crystal structure of the TTD in complex with 5A-DMP revealed that the compound stably bound to the Arg-binding cavity of the TTD. Furthermore, 5A-DMP inhibits the full-length UHRF1:LIG1 interaction in Xenopus egg extracts. Our study uncovers a UHRF1 inhibitor which can be the basis of future experiments for cancer therapy.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , ADN Ligasa (ATP)/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Simulación de Dinámica Molecular , Piridinas/farmacología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular Tumoral , ADN Ligasa (ATP)/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Piridinas/química , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Xenopus
7.
Nucleic Acids Res ; 47(7): 3407-3421, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30753595

RESUMEN

The proper tissue-specific regulation of gene expression is essential for development and homeostasis in metazoans. However, the illegitimate expression of normally tissue-restricted genes-like testis- or placenta-specific genes-is frequently observed in tumors; this promotes transformation, but also allows immunotherapy. Two important questions are: how is the expression of these genes controlled in healthy cells? And how is this altered in cancer? To address these questions, we used an unbiased approach to test the ability of 350 distinct genetic or epigenetic perturbations to induce the illegitimate expression of over 40 tissue-restricted genes in primary human cells. We find that almost all of these genes are remarkably resistant to reactivation by a single alteration in signaling pathways or chromatin regulation. However, a few genes differ and are more readily activated; one is the placenta-expressed gene ADAM12, which promotes invasion. Using cellular systems, an animal model, and bioinformatics, we find that a non-canonical but druggable TGF-ß/KAT2A/TAK1 axis controls ADAM12 induction in normal and cancer cells. More broadly, our data show that illegitimate gene expression in cancer is an heterogeneous phenomenon, with a few genes activatable by simple events, and most genes likely requiring a combination of events to become reactivated.


Asunto(s)
Regulación de la Expresión Génica/genética , Neoplasias/genética , Especificidad de Órganos/genética , Transcripción Genética/genética , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Histona Acetiltransferasas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Factor de Crecimiento Transformador beta1/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
8.
Nucleic Acids Res ; 46(9): 4392-4404, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29490077

RESUMEN

Reactive oxygen species (ROS) are a byproduct of cell metabolism, and can also arise from environmental sources, such as toxins or radiation. Depending on dose and context, ROS have both beneficial and deleterious roles in mammalian development and disease, therefore it is crucial to understand how these molecules are generated, sensed, and detoxified. The question of how oxidative stress connects to the epigenome, in particular, is important yet incompletely understood. Here we show that an epigenetic regulator, the methyl-CpG-binding protein ZBTB38, limits the basal cellular production of ROS, is induced by ROS, and is required to mount a proper response to oxidative stress. Molecularly, these functions depend on a deubiquitinase, USP9X, which interacts with ZBTB38, deubiquitinates it, and stabilizes it. We find that USP9X is itself stabilized by oxidative stress, and is required together with ZBTB38 to limit the basal generation of ROS, as well as the toxicity of an acute oxidative stress. Our data uncover a new nuclear target of USP9X, show that the USP9X/ZBTB38 axis limits, senses and detoxifies ROS, and provide a molecular link between oxidative stress and the epigenome.


Asunto(s)
Estrés Oxidativo , Proteínas Represoras/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Línea Celular Tumoral , Núcleo Celular/enzimología , Núcleo Celular/metabolismo , Humanos , Estabilidad Proteica , Especies Reactivas de Oxígeno/metabolismo
9.
Nucleic Acids Res ; 44(18): 8621-8640, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27302134

RESUMEN

Myogenic regulatory factors of the MyoD family have the ability to reprogram differentiated cells toward a myogenic fate. In this study, we demonstrate that Six1 or Six4 are required for the reprogramming by MyoD of mouse embryonic fibroblasts (MEFs). Using microarray experiments, we found 761 genes under the control of both Six and MyoD. Using MyoD ChIPseq data and a genome-wide search for Six1/4 MEF3 binding sites, we found significant co-localization of binding sites for MyoD and Six proteins on over a thousand mouse genomic DNA regions. The combination of both datasets yielded 82 genes which are synergistically activated by Six and MyoD, with 96 associated MyoD+MEF3 putative cis-regulatory modules (CRMs). Fourteen out of 19 of the CRMs that we tested demonstrated in Luciferase assays a synergistic action also observed for their cognate gene. We searched putative binding sites on these CRMs using available databases and de novo search of conserved motifs and demonstrated that the Six/MyoD synergistic activation takes place in a feedforward way. It involves the recruitment of these two families of transcription factors to their targets, together with partner transcription factors, encoded by genes that are themselves activated by Six and MyoD, including Mef2, Pbx-Meis and EBF.


Asunto(s)
Reprogramación Celular/genética , Genoma , Proteínas de Homeodominio/metabolismo , Proteína MioD/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transactivadores/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Proteínas de Homeodominio/genética , Humanos , Luciferasas/metabolismo , Ratones Noqueados , Desarrollo de Músculos/genética , Mutación/genética , Proteínas Nucleares/metabolismo , Motivos de Nucleótidos/genética , Reproducibilidad de los Resultados , Transactivadores/genética , Factores de Transcripción/metabolismo
10.
Proteomics ; 14(19): 2179-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24634419

RESUMEN

MBD5 and MBD6 are two members of the methyl-CpG-binding domain (MBD) family of proteins that are poorly characterized. Studies performed thus far have failed to show binding of the MBD5 and MBD6 MBD to methylated DNA. Here, we show that both MBD5 and MBD6 interact with the mammalian PR-DUB Polycomb protein complex in a mutually exclusive manner. Strikingly, the MBD of MBD5 and MBD6 is both necessary and sufficient to mediate this interaction. Chromatin immunoprecipitation analyses reveal that MBD6 and FOXK2/PR-DUB share a subset of genomic target genes, suggesting a functional interaction in vivo. Finally, we show that MBD6, but not MBD5, is recruited to sites of DNA damage in a PR-DUB independent manner. Our study thus implies a shared function for MBD5 and MBD6 through an interaction with PR-DUB, as well as an MBD6-specific recruitment to sites of DNA damage.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Secuencia de Aminoácidos , Cromatina , Daño del ADN , Metilación de ADN , Factores de Transcripción Forkhead , Células HEK293 , Células HeLa , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo
11.
Med Sci (Paris) ; 40(2): 147-153, 2024 Feb.
Artículo en Francés | MEDLINE | ID: mdl-38411422

RESUMEN

Totipotency is the ability of a cell to generate a whole organism, a property that characterizes the first embryonic cells, such as the zygote and the blastomeres. This review provides a retrospective on the progress made in the last decade in the study of totipotency, especially with the discovery of mouse ES cells expressing markers of the 2-cell stage (2C-like cells). This model has greatly contributed to a better understanding of the molecular mechanisms involved in totipotency (pioneer factors, epigenetic regulation, splicing, nuclear maturation). 2C-like cells have also paved the way for the development of new cellular models of human totipotency.


Title: Comprendre la totipotence embryonnaire à partir des cellules 2C-like. Abstract: La totipotence est la capacité d'une cellule à générer un organisme entier, une propriété qui caractérise les premières cellules embryonnaires, comme le zygote et les blastomères. Dans cette revue, nous proposons une rétrospective des avancées réalisées au cours de la dernière décennie concernant l'étude de la totipotence avec, notamment, la découverte des cellules ES murines exprimant des marqueurs du stade 2-cellules (2CLC). Ce modèle a considérablement contribué à la meilleure compréhension des mécanismes moléculaires impliqués dans la totipotence (facteurs pionniers, régulation épigénétique, épissage, maturation nucléaire). Les cellules 2CLC ont aussi ouvert la voie au développement de nouveaux modèles cellulaires de totipotence humaine.


Asunto(s)
Células Madre Embrionarias , Epigénesis Genética , Humanos , Animales , Ratones , Estudios Retrospectivos , Empalme del ARN , Cigoto
12.
Cell Genom ; 4(2): 100498, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38309261

RESUMEN

Long interspersed element 1 (L1) retrotransposons are implicated in human disease and evolution. Their global activity is repressed by DNA methylation, but deciphering the regulation of individual copies has been challenging. Here, we combine short- and long-read sequencing to unveil L1 methylation heterogeneity across cell types, families, and individual loci and elucidate key principles involved. We find that the youngest primate L1 families are specifically hypomethylated in pluripotent stem cells and the placenta but not in most tumors. Locally, intronic L1 methylation is intimately associated with gene transcription. Conversely, the L1 methylation state can propagate to the proximal region up to 300 bp. This phenomenon is accompanied by the binding of specific transcription factors, which drive the expression of L1 and chimeric transcripts. Finally, L1 hypomethylation alone is typically insufficient to trigger L1 expression due to redundant silencing pathways. Our results illuminate the epigenetic and transcriptional interplay between retrotransposons and their host genome.


Asunto(s)
Metilación de ADN , Retroelementos , Animales , Humanos , Retroelementos/genética , Metilación de ADN/genética , Elementos de Nucleótido Esparcido Largo/genética , Factores de Transcripción/genética , Primates/genética , Epigénesis Genética/genética
13.
Oncogene ; 43(18): 1369-1385, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38467851

RESUMEN

Breast cancer is the most prevalent type of cancer in women worldwide. Within breast tumors, the basal-like subtype has the worst prognosis, prompting the need for new tools to understand, detect, and treat these tumors. Certain germline-restricted genes show aberrant expression in tumors and are known as Cancer/Testis genes; their misexpression has diagnostic and therapeutic applications. Here we designed a new bioinformatic approach to examine Cancer/Testis gene misexpression in breast tumors. We identify several new markers in Luminal and HER-2 positive tumors, some of which predict response to chemotherapy. We then use machine learning to identify the two Cancer/Testis genes most associated with basal-like breast tumors: HORMAD1 and CT83. We show that these genes are expressed by tumor cells and not by the microenvironment, and that they are not expressed by normal breast progenitors; in other words, their activation occurs de novo. We find these genes are epigenetically repressed by DNA methylation, and that their activation upon DNA demethylation is irreversible, providing a memory of past epigenetic disturbances. Simultaneous expression of both genes in breast cells in vitro has a synergistic effect that increases stemness and activates a transcriptional profile also observed in double-positive tumors. Therefore, we reveal a functional cooperation between Cancer/Testis genes in basal breast tumors; these findings have consequences for the understanding, diagnosis, and therapy of the breast tumors with the worst outcomes.


Asunto(s)
Neoplasias de la Mama , Biología Computacional , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Biología Computacional/métodos , Metilación de ADN , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Masculino , Epigénesis Genética
14.
Nat Commun ; 15(1): 2960, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580649

RESUMEN

DNA methylation is an essential epigenetic chromatin modification, and its maintenance in mammals requires the protein UHRF1. It is yet unclear if UHRF1 functions solely by stimulating DNA methylation maintenance by DNMT1, or if it has important additional functions. Using degron alleles, we show that UHRF1 depletion causes a much greater loss of DNA methylation than DNMT1 depletion. This is not caused by passive demethylation as UHRF1-depleted cells proliferate more slowly than DNMT1-depleted cells. Instead, bioinformatics, proteomics and genetics experiments establish that UHRF1, besides activating DNMT1, interacts with DNMT3A and DNMT3B and promotes their activity. In addition, we show that UHRF1 antagonizes active DNA demethylation by TET2. Therefore, UHRF1 has non-canonical roles that contribute importantly to DNA methylation homeostasis; these findings have practical implications for epigenetics in health and disease.


Asunto(s)
Metilación de ADN , Neoplasias , Humanos , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Cromatina , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Neoplasias/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
15.
Sci Rep ; 13(1): 4363, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36928068

RESUMEN

DNA ligase I (LigI), the predominant enzyme that joins Okazaki fragments, interacts with PCNA and Pol δ. LigI also interacts with UHRF1, linking Okazaki fragment joining with DNA maintenance methylation. Okazaki fragments can also be joined by a relatively poorly characterized DNA ligase IIIα (LigIIIα)-dependent backup pathway. Here we examined the effect of LigI-deficiency on proteins at the replication fork. Notably, LigI-deficiency did not alter the kinetics of association of the PCNA clamp, the leading strand polymerase Pol ε, DNA maintenance methylation proteins and core histones with newly synthesized DNA. While the absence of major changes in replication and methylation proteins is consistent with the similar proliferation rate and DNA methylation levels of the LIG1 null cells compared with the parental cells, the increased levels of LigIIIα/XRCC1 and Pol δ at the replication fork and in bulk chromatin indicate that there are subtle replication defects in the absence of LigI. Interestingly, the non-replicative histone H1 variant, H1.0, is enriched in the chromatin of LigI-deficient mouse CH12F3 and human 46BR.1G1 cells. This alteration was not corrected by expression of wild type LigI, suggesting that it is a relatively stable epigenetic change that may contribute to the immunodeficiencies linked with inherited LigI-deficiency syndrome.


Asunto(s)
ADN Ligasa (ATP) , Replicación del ADN , Histonas , Antígeno Nuclear de Célula en Proliferación , Animales , Humanos , Ratones , Cromatina/genética , ADN/metabolismo , ADN Ligasa (ATP)/genética , ADN Ligasa (ATP)/metabolismo , ADN Ligasas/genética , ADN Ligasas/metabolismo , ADN Polimerasa III/genética , Histonas/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo
16.
J Exp Clin Cancer Res ; 42(1): 318, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38008756

RESUMEN

BACKGROUND: TGFß induces several cell phenotypes including senescence, a stable cell cycle arrest accompanied by a secretory program, and epithelial-mesenchymal transition (EMT) in normal epithelial cells. During carcinogenesis cells lose the ability to undergo senescence in response to TGFß but they maintain an EMT, which can contribute to tumor progression. Our aim was to identify mechanisms promoting TGFß-induced senescence escape. METHODS: In vitro experiments were performed with primary human mammary epithelial cells (HMEC) immortalized by hTert. For kinase library screen and modulation of gene expression retroviral transduction was used. To characterize gene expression, RNA microarray with GSEA analysis and RT-qPCR were used. For protein level and localization, Western blot and immunofluorescence were performed. For senescence characterization crystal violet assay, Senescence Associated-ß-Galactosidase activity, EdU staining were conducted. To determine RSK3 partners FLAG-baited immunoprecipitation and mass spectrometry-based proteomic analyses were performed. Proteosome activity and proteasome enrichment assays were performed. To validate the role of RSK3 in human breast cancer, analysis of METABRIC database was performed. Murine intraductal xenografts using MCF10DCIS.com cells were carried out, with histological and immunofluorescence analysis of mouse tissue sections. RESULTS: A screen with active kinases in HMECs upon TGFß treatment identified that the serine threonine kinase RSK3, or RPS6KA2, a kinase mainly known to regulate cancer cell death including in breast cancer, reverted TGFß-induced senescence. Interestingly, RSK3 expression decreased in response to TGFß in a SMAD3-dependent manner, and its constitutive expression rescued SMAD3-induced senescence, indicating that a decrease in RSK3 itself contributes to TGFß-induced senescence. Using transcriptomic analyses and affinity purification coupled to mass spectrometry-based proteomics, we unveiled that RSK3 regulates senescence by inhibiting the NF-κΒ pathway through the decrease in proteasome-mediated IκBα degradation. Strikingly, senescent TGFß-treated HMECs display features of epithelial to mesenchymal transition (EMT) and during RSK3-induced senescence escaped HMECs conserve EMT features. Importantly, RSK3 expression is correlated with EMT and invasion, and inversely correlated with senescence and NF-κΒ in human claudin-low breast tumors and its expression enhances the formation of breast invasive tumors in the mouse mammary gland. CONCLUSIONS: We conclude that RSK3 switches cell fate from senescence to malignancy in response to TGFß signaling.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/patología , Transición Epitelial-Mesenquimal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
17.
Nat Struct Mol Biol ; 30(8): 1105-1118, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37488355

RESUMEN

In mammals, only the zygote and blastomeres of the early embryo are totipotent. This totipotency is mirrored in vitro by mouse '2-cell-like cells' (2CLCs), which appear at low frequency in cultures of embryonic stem cells (ESCs). Because totipotency is not completely understood, we carried out a genome-wide CRISPR knockout screen in mouse ESCs, searching for mutants that reactivate the expression of Dazl, a gene expressed in 2CLCs. Here we report the identification of four mutants that reactivate Dazl and a broader 2-cell-like signature: the E3 ubiquitin ligase adaptor SPOP, the Zinc-Finger transcription factor ZBTB14, MCM3AP, a component of the RNA processing complex TREX-2, and the lysine demethylase KDM5C. All four factors function upstream of DPPA2 and DUX, but not via p53. In addition, SPOP binds DPPA2, and KDM5C interacts with ncPRC1.6 and inhibits 2CLC gene expression in a catalytic-independent manner. These results extend our knowledge of totipotency, a key phase of organismal life.


Asunto(s)
Factores de Transcripción , Cigoto , Ratones , Animales , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Madre Embrionarias/metabolismo , Genoma , Células Madre Embrionarias de Ratones/metabolismo , Mamíferos/genética
18.
EMBO J ; 27(11): 1563-74, 2008 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-18451802

RESUMEN

In response to stimuli that activate p53, cells can undergo either apoptosis or cell cycle arrest, depending on the precise pattern of p53 target genes that is activated. We show here that Zbtb4, a transcriptional repressor protein, associates with the Sin3/histone deacetylase co-repressor and represses expression of P21CIP1 as part of a heterodimeric complex with Miz1. In vivo, expression of ZBTB4 is downregulated in advanced stages of multiple human tumours. In cell culture, depletion of ZBTB4 promotes cell cycle arrest in response to activation of p53 and suppresses apoptosis through regulation of P21CIP1, thereby promoting long-term cell survival. Our data suggest that Zbtb4 is a critical determinant of the cellular response to p53 activation and reinforce the notion that p21Cip1 can provide an essential survival signal in cells with activated p53.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Ciclo Celular , Niño , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Complejo Correpresor Histona Desacetilasa y Sin3 , Factores de Transcripción/metabolismo , Transcripción Genética , Células Tumorales Cultivadas
19.
Bioessays ; 32(12): 1025-32, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20886526

RESUMEN

CpG islands (CGIs) are regions enriched in the dinucleotide CpG; they constitute the promoter of about 60% of mammalian genes. In cancer cells, some promoter-associated CGIs become heavily methylated on cytosines, and the corresponding genes undergo stable transcriptional silencing. Hypermethylated CGIs attract methyl-CpG-binding proteins (MBPs), which have been shown to recruit chromatin modifiers and cause transcriptional repression. These observations have led to the prevalent model that methyl-CpG-binding proteins are promoter-proximal transcriptional repressors. Recent discoveries challenge this idea and raise a number of questions. Here we discuss the following issues: what are other possible roles for the known MBPs? Why are these proteins not essential in mammals? Are there other MBPs left to discover? Could CpG methylation be nonessential?


Asunto(s)
Islas de CpG , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Proteína 2 de Unión a Metil-CpG/metabolismo , Regiones Promotoras Genéticas , Animales , Ensamble y Desensamble de Cromatina , Citosina , Proteínas de Unión al ADN/genética , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Elementos Silenciadores Transcripcionales , Transcripción Genética
20.
Nucleic Acids Res ; 38(15): 5015-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20403812

RESUMEN

DNA methylation is an essential epigenetic mark. Three classes of mammalian proteins recognize methylated DNA: MBD proteins, SRA proteins and the zinc-finger proteins Kaiso, ZBTB4 and ZBTB38. The last three proteins can bind either methylated DNA or unmethylated consensus sequences; how this is achieved is largely unclear. Here, we report that the human zinc-finger proteins Kaiso, ZBTB4 and ZBTB38 can bind methylated DNA in a sequence-specific manner, and that they may use a mode of binding common to other zinc-finger proteins. This suggests that many other sequence-specific methyl binding proteins may exist.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Bases , Sitios de Unión , Secuencia de Consenso , Proteínas de Unión al ADN/química , Humanos , Unión Proteica , Proteínas Represoras/química , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA