Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO J ; 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951609

RESUMEN

Transposable elements (TEs) are mobile genetic modules of viral derivation that have been co-opted to become modulators of mammalian gene expression. TEs are a major source of endogenous dsRNAs, signaling molecules able to coordinate inflammatory responses in various physiological processes. Here, we provide evidence for a positive involvement of TEs in inflammation-driven bone repair and mineralization. In newly fractured mice bone, we observed an early transient upregulation of repeats occurring concurrently with the initiation of the inflammatory stage. In human bone biopsies, analysis revealed a significant correlation between repeats expression, mechanical stress and bone mineral density. We investigated a potential link between LINE-1 (L1) expression and bone mineralization by delivering a synthetic L1 RNA to osteoporotic patient-derived mesenchymal stem cells and observed a dsRNA-triggered protein kinase (PKR)-mediated stress response that led to strongly increased mineralization. This response was associated with a strong and transient inflammation, accompanied by a global translation attenuation induced by eIF2α phosphorylation. We demonstrated that L1 transfection reshaped the secretory profile of osteoblasts, triggering a paracrine activity that stimulated the mineralization of recipient cells.

2.
Cell Rep ; 42(9): 113066, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37656620

RESUMEN

Fear-related disorders arise from inefficient fear extinction and have immeasurable social and economic costs. Here, we characterize mouse phenotypes that spontaneously show fear-independent behavioral traits predicting adaptive or maladaptive fear extinction. We find that, already before fear conditioning, specific morphological, electrophysiological, and transcriptomic patterns of cortical and amygdala pyramidal neurons predispose to fear-related disorders. Finally, by using an optogenetic approach, we show the possibility to rescue inefficient fear extinction by activating infralimbic pyramidal neurons and to impair fear extinction by activating prelimbic pyramidal neurons.


Asunto(s)
Miedo , Corteza Prefrontal , Ratones , Animales , Corteza Prefrontal/fisiología , Miedo/fisiología , Transcriptoma/genética , Extinción Psicológica/fisiología , Amígdala del Cerebelo/fisiología , Células Piramidales/fisiología
3.
Int J Mol Sci ; 24(18)2023 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-37762112

RESUMEN

Since its initial involvement in numerous neurodegenerative pathologies in 2006, either as a principal actor or as a cofactor, new pathologies implicating transactive response (TAR) DNA-binding protein 43 (TDP-43) are regularly emerging also beyond the neuronal system. This reflects the fact that TDP-43 functions are particularly complex and broad in a great variety of human cells. In neurodegenerative diseases, this protein is often pathologically delocalized to the cytoplasm, where it irreversibly aggregates and is subjected to various post-translational modifications such as phosphorylation, polyubiquitination, and cleavage. Until a few years ago, the research emphasis has been focused particularly on the impacts of this aggregation and/or on its widely described role in complex RNA splicing, whether related to loss- or gain-of-function mechanisms. Interestingly, recent studies have strengthened the knowledge of TDP-43 activity at the chromatin level and its implication in the regulation of DNA transcription and stability. These discoveries have highlighted new features regarding its own transcriptional regulation and suggested additional mechanistic and disease models for the effects of TPD-43. In this review, we aim to give a comprehensive view of the potential epigenetic (de)regulations driven by (and driving) this multitask DNA/RNA-binding protein.


Asunto(s)
Cromatina , Proteínas de Unión al ADN , Humanos , Citoplasma , Proteínas de Unión al ADN/genética , Epigénesis Genética , Epigenómica
5.
Cell Rep ; 42(6): 112588, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37267101

RESUMEN

Physiology is regulated by interconnected cell and tissue circadian clocks. Disruption of the rhythms generated by the concerted activity of these clocks is associated with metabolic disease. Here we tested the interactions between clocks in two critical components of organismal metabolism, liver and skeletal muscle, by rescuing clock function either in each organ separately or in both organs simultaneously in otherwise clock-less mice. Experiments showed that individual clocks are partially sufficient for tissue glucose metabolism, yet the connections between both tissue clocks coupled to daily feeding rhythms support systemic glucose tolerance. This synergy relies in part on local transcriptional control of the glucose machinery, feeding-responsive signals such as insulin, and metabolic cycles that connect the muscle and liver. We posit that spatiotemporal mechanisms of muscle and liver play an essential role in the maintenance of systemic glucose homeostasis and that disrupting this diurnal coordination can contribute to metabolic disease.


Asunto(s)
Relojes Circadianos , Ratones , Animales , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Hígado/metabolismo , Músculo Esquelético/metabolismo , Glucosa/metabolismo
6.
Methods Mol Biol ; 2655: 31-39, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37212986

RESUMEN

Upon cellular reprogramming, the activity of polycomb repressive complex 2 (PRC2), together with histone demethylases, is essential for the suppression of cell lineage-specific gene expression programs, for resetting of epigenetic memory and for the reacquisition of pluripotency.PRC2 requires interaction with RNAs for the correct protein complex assembly and recruitment on chromatin. Moreover, PRC2 components can be found in different cell compartments and their intracellular dynamics is part of their functional activity. Several loss-of-function studies revealed that many lncRNAs expressed upon reprogramming are essential for the silencing of lineage-specific genes and the function of chromatin modifiers. Compartment-specific UV-RIP technique is a method that will help understanding which is the nature of those interactions, with no interference from indirect interactions typical of methods involving the use of chemical cross-linkers or performed in native conditions with non-stringent buffers. This technique will shed lights on the specificity of lncRNA interaction and PRC2 stability/activity on chromatin and whether PRC2-lncRNA interaction occurs in specific cell compartments.


Asunto(s)
Complejo Represivo Polycomb 2 , ARN Largo no Codificante , Complejo Represivo Polycomb 2/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromatina/genética
7.
Methods Mol Biol ; 2655: 101-116, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37212992

RESUMEN

The Polycomb repressive complex 2 (PRC2) is a well-characterized chromatin regulator of transcription programs acting through H3K27me3 deposition. In mammals, there are two main versions of PRC2 complexes: PRC2-EZH2, which is prevalent in cycling cells, and PRC2-EZH1 where EZH1 replaces EZH2 in post-mitotic tissues. Stoichiometry of PRC2 complex is dynamically modulated during cellular differentiation and various stress conditions. Therefore, unraveling unique architecture of PRC2 complexes under specific biological context through comprehensive and quantitative characterization could provide insight into the underlying mechanistic molecular mechanism in regulation of transcription process. In this chapter, we describe an efficient method which combines tandem-affinity purification (TAP) with label-free quantitative proteomics strategy for studying PRC2-EZH1 complex architecture alterations and identifying novel protein regulators in post-mitotic C2C12 skeletal muscle cells.


Asunto(s)
Histonas , Complejo Represivo Polycomb 2 , Animales , Complejo Represivo Polycomb 2/genética , Histonas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Cromatina , Mamíferos/metabolismo
8.
J Vis Exp ; (193)2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-37010313

RESUMEN

Histone post-translational modifications (PTMs) and other epigenetic modifications regulate the chromatin accessibility of genes to the transcriptional machinery, thus affecting an organism's capacity to respond to environmental stimuli. Chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) has been widely utilized to identify and map protein-DNA interactions in the fields of epigenetics and gene regulation. However, the field of cnidarian epigenetics is hampered by a lack of applicable protocols, partly due to the unique features of model organisms such as the symbiotic sea anemone Exaiptasia diaphana, whose high water content and mucus amounts obstruct molecular methods. Here, a specialized ChIP procedure is presented, which facilitates the investigation of protein-DNA interactions in E. diaphana gene regulation. The cross-linking and chromatin extraction steps were optimized for efficient immunoprecipitation and then validated by performing ChIP using an antibody against the histone mark H3K4me3. Subsequently, the specificity and effectiveness of the ChIP assay were confirmed by measuring the relative occupancy of H3K4me3 around several constitutively activated gene loci using quantitative PCR and by next-generation sequencing for genome-wide scale analysis. This optimized ChIP protocol for the symbiotic sea anemone E. diaphana facilitates the investigation of the protein-DNA interactions involved in organismal responses to environmental changes that affect symbiotic cnidarians, such as corals.


Asunto(s)
Anémonas de Mar , Animales , Anémonas de Mar/genética , Cromatina/genética , Inmunoprecipitación de Cromatina/métodos , Secuenciación de Inmunoprecipitación de Cromatina/métodos , ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos
9.
Front Cell Dev Biol ; 11: 1123975, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36760365

RESUMEN

Non-coding DNA accounts for approximately 98.5% of the human genome. Once labeled as "junk DNA", this portion of the genome has undergone a progressive re-evaluation and it is now clear that some of its transcriptional products, belonging to the non-coding RNAs (ncRNAs), are key players in cell regulatory networks. A growing body of evidence demonstrates the crucial impact of regulatory ncRNAs on mammalian gene expression. Here, we focus on the defined relationship between chromatin-interacting RNAs, particularly long non-coding RNA (lncRNA), enhancer RNA (eRNA), non-coding natural antisense transcript (ncNAT), and circular RNA (circRNA) and epigenome, a common ground where both protein and RNA species converge to regulate cellular functions. Through several examples, this review provides an overview of the variety of targets, interactors, and mechanisms involved in the RNA-mediated modulation of loci-specific epigenetic states, a fundamental evolutive strategy to orchestrate mammalian gene expression in a timely and reversible manner. We will discuss how RNA-mediated epigenetic regulation impacts development and tissue homeostasis and how its alteration contributes to the onset and progression of many different human diseases, particularly cancer.

10.
Sci Transl Med ; 14(657): eabl6057, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947677

RESUMEN

Constitutive heterochromatin is responsible for genome repression of DNA enriched in repetitive sequences, telomeres, and centromeres. During physiological and pathological premature aging, heterochromatin homeostasis is profoundly compromised. Here, we showed that LINE-1 (Long Interspersed Nuclear Element-1; L1) RNA accumulation was an early event in both typical and atypical human progeroid syndromes. L1 RNA negatively regulated the enzymatic activity of the histone-lysine N-methyltransferase SUV39H1 (suppression of variegation 3-9 homolog 1), resulting in heterochromatin loss and onset of senescent phenotypes in vitro. Depletion of L1 RNA in dermal fibroblast cells from patients with different progeroid syndromes using specific antisense oligonucleotides (ASOs) restored heterochromatin histone 3 lysine 9 and histone 3 lysine 27 trimethylation marks, reversed DNA methylation age, and counteracted the expression of senescence-associated secretory phenotype genes such as p16, p21, activating transcription factor 3 (ATF3), matrix metallopeptidase 13 (MMP13), interleukin 1a (IL1a), BTG anti-proliferation factor 2 (BTG2), and growth arrest and DNA damage inducible beta (GADD45b). Moreover, systemic delivery of ASOs rescued the histophysiology of tissues and increased the life span of a Hutchinson-Gilford progeria syndrome mouse model. Transcriptional profiling of human and mouse samples after L1 RNA depletion demonstrated that pathways associated with nuclear chromatin organization, cell proliferation, and transcription regulation were enriched. Similarly, pathways associated with aging, inflammatory response, innate immune response, and DNA damage were down-regulated. Our results highlight the role of L1 RNA in heterochromatin homeostasis in progeroid syndromes and identify a possible therapeutic approach to treat premature aging and related syndromes.


Asunto(s)
Envejecimiento Prematuro , Síndrome de Cockayne , Proteínas Inmediatas-Precoces , Progeria , Envejecimiento Prematuro/genética , Animales , Antígenos de Diferenciación , Heterocromatina , Histonas/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Elementos de Nucleótido Esparcido Largo , Lisina/metabolismo , Ratones , Fenotipo , Progeria/genética , ARN , Telómero/genética , Proteínas Supresoras de Tumor/genética
11.
Int J Mol Sci ; 23(5)2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35269678

RESUMEN

Approach and avoidance (A/A) tendencies are stable behavioral traits in responding to rewarding and fearful stimuli. They represent the superordinate division of emotion, and individual differences in such traits are associated with disease susceptibility. The neural circuitry underlying A/A traits is retained to be the cortico-limbic pathway including the amygdala, the central hub for the emotional processing. Furthermore, A/A-specific individual differences are associated with the activity of the endocannabinoid system (ECS) and especially of CB1 receptors whose density and functionality in amygdala differ according to A/A traits. ECS markedly interacts with the immune system (IS). However, how the interplay between ECS and IS is associated with A/A individual differences is still ill-defined. To fill this gap, here we analyzed the interaction between the gene expression of ECS and immune system (IS) in relation to individual differences. To unveil the deep architecture of ECS-IS interaction, we performed cell-specific transcriptomics analysis. Differential gene expression profiling, functional enrichment, and protein-protein interaction network analyses were performed in amygdala pyramidal neurons of mice showing different A/A behavioral tendencies. Several altered pro-inflammatory pathways were identified as associated with individual differences in A/A traits, indicating the chronic activation of the adaptive immune response sustained by the interplay between endocannabinoids and the IS. Furthermore, results showed that the interaction between the two systems modulates synaptic plasticity and neuronal metabolism in individual difference-specific manner. Deepening our knowledge about ECS/IS interaction may provide useful targets for treatment and prevention of psychopathology associated with A/A traits.


Asunto(s)
Endocannabinoides , Transcriptoma , Amígdala del Cerebelo/metabolismo , Animales , Endocannabinoides/metabolismo , Ratones , Plasticidad Neuronal , Neuronas/metabolismo , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo
12.
Cell Rep ; 37(9): 110066, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34852230

RESUMEN

The role of chromatin-associated RNAi components in the nucleus of mammalian cells and in particular in the context of developmental programs remains to be elucidated. Here, we investigate the function of nuclear Argonaute 1 (Ago1) in gene expression regulation during skeletal muscle differentiation. We show that Ago1 is required for activation of the myogenic program by supporting chromatin modification mediated by developmental enhancer activation. Mechanistically, we demonstrate that Ago1 directly controls global H3K27 acetylation (H3K27ac) by regulating enhancer RNA (eRNA)-CREB-binding protein (CBP) acetyltransferase interaction, a key step in enhancer-driven gene activation. In particular, we show that Ago1 is specifically required for myogenic differentiation 1 (MyoD) and downstream myogenic gene activation, whereas its depletion leads to failure of CBP acetyltransferase activation and blocking of the myogenic program. Our work establishes a role of the mammalian enhancer-associated RNAi component Ago1 in epigenome regulation and activation of developmental programs.


Asunto(s)
Proteínas Argonautas/metabolismo , Epigenoma , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Proteínas de la Membrana/metabolismo , Mioblastos/citología , Fosfoproteínas/metabolismo , ARN no Traducido/metabolismo , Acetilación , Animales , Proteínas Argonautas/genética , Diferenciación Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Factores Eucarióticos de Iniciación/genética , Histonas/genética , Proteínas de la Membrana/genética , Ratones , Desarrollo de Músculos , Mioblastos/metabolismo , Fosfoproteínas/genética , ARN no Traducido/genética , Transcripción Genética
13.
Cell Death Dis ; 12(10): 850, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34531374

RESUMEN

PRC2-mediated epigenetic function involves the interaction with long non-coding RNAs (lncRNAs). Although the identity of some of these RNAs has been elucidated in the context of developmental programs, their counterparts in postmitotic adult tissue homeostasis remain uncharacterized. To this aim, we used terminally differentiated postmitotic skeletal muscle cells in which oxidative stress induces the dynamic activation of PRC2-Ezh1 through Embryonic Ectoderm Develpment (EED) shuttling to the nucleus. We identify lncRNA Malat-1 as a necessary partner for PRC2-Ezh1-dependent response to oxidative stress. We show that in this pathway, PRC2-EZH1 dynamic assembly, and in turn stress induced skeletal muscle targeted genes repression, depends specifically on Malat-1. Our study reports about PRC2-RNA interactions in the physiological context of adaptive oxidative stress response and identifies the first lncRNA involved in PRC2-Ezh1 function.


Asunto(s)
Epigenoma , Fibras Musculares Esqueléticas/metabolismo , Estrés Oxidativo , Complejo Represivo Polycomb 2/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Línea Celular , Ectodermo/embriología , Embrión de Mamíferos/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Modelos Biológicos , Atrofia Muscular/genética , Atrofia Muscular/patología , Estrés Oxidativo/genética , Fenotipo , Complejo Represivo Polycomb 2/genética , Unión Proteica , ARN Largo no Codificante/genética , Transcripción Genética
15.
Cell Mol Life Sci ; 78(13): 5245-5256, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33990851

RESUMEN

Retrotransposons are genetic elements present across all eukaryotic genomes. While their role in evolution is considered as a potentially beneficial natural source of genetic variation, their activity is classically considered detrimental due to their potentially harmful effects on genome stability. However, studies are increasingly shedding light on the regulatory function and beneficial role of somatic retroelement reactivation in non-pathological contexts. Here, we review recent findings unveiling the regulatory potential of retrotransposons, including their role in noncoding RNA transcription, as modulators of mammalian transcriptional and epigenome landscapes. We also discuss technical challenges in deciphering the multifaceted activity of retrotransposable elements, highlighting an unforeseen central role of this neglected portion of the genome both in early development and in adult life.


Asunto(s)
Epigenoma , Evolución Molecular , Inestabilidad Genómica , Mamíferos/genética , ARN no Traducido/genética , Retroelementos , Animales , Humanos , Mamíferos/crecimiento & desarrollo
16.
Int J Mol Sci ; 22(2)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467450

RESUMEN

Fear extinction requires coordinated neural activity within the amygdala and medial prefrontal cortex (mPFC). Any behavior has a transcriptomic signature that is modified by environmental experiences, and specific genes are involved in functional plasticity and synaptic wiring during fear extinction. Here, we investigated the effects of optogenetic manipulations of prelimbic (PrL) pyramidal neurons and amygdala gene expression to analyze the specific transcriptional pathways associated to adaptive and maladaptive fear extinction. To this aim, transgenic mice were (or not) fear-conditioned and during the extinction phase they received optogenetic (or sham) stimulations over photo-activable PrL pyramidal neurons. At the end of behavioral testing, electrophysiological (neural cellular excitability and Excitatory Post-Synaptic Currents) and morphological (spinogenesis) correlates were evaluated in the PrL pyramidal neurons. Furthermore, transcriptomic cell-specific RNA-analyses (differential gene expression profiling and functional enrichment analyses) were performed in amygdala pyramidal neurons. Our results show that the optogenetic activation of PrL pyramidal neurons in fear-conditioned mice induces fear extinction deficits, reflected in an increase of cellular excitability, excitatory neurotransmission, and spinogenesis of PrL pyramidal neurons, and associated to strong modifications of the transcriptome of amygdala pyramidal neurons. Understanding the electrophysiological, morphological, and transcriptomic architecture of fear extinction may facilitate the comprehension of fear-related disorders.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Células Piramidales/fisiología , Transcriptoma/genética , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/metabolismo , Animales , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores/fisiología , Miedo/psicología , Masculino , Memoria/fisiología , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiología , Optogenética/métodos , Corteza Prefrontal/citología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Células Piramidales/metabolismo , Transmisión Sináptica/fisiología
17.
Nat Commun ; 11(1): 1018, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094342

RESUMEN

Mammalian genomes encode tens of thousands of noncoding RNAs. Most noncoding transcripts exhibit nuclear localization and several have been shown to play a role in the regulation of gene expression and chromatin remodeling. To investigate the function of such RNAs, methods to massively map the genomic interacting sites of multiple transcripts have been developed; however, these methods have some limitations. Here, we introduce RNA And DNA Interacting Complexes Ligated and sequenced (RADICL-seq), a technology that maps genome-wide RNA-chromatin interactions in intact nuclei. RADICL-seq is a proximity ligation-based methodology that reduces the bias for nascent transcription, while increasing genomic coverage and unique mapping rate efficiency compared with existing methods. RADICL-seq identifies distinct patterns of genome occupancy for different classes of transcripts as well as cell type-specific RNA-chromatin interactions, and highlights the role of transcription in the establishment of chromatin structure.


Asunto(s)
Cromatina/metabolismo , Mapeo Cromosómico/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ARN no Traducido/genética , Análisis de Secuencia de ARN/métodos , Animales , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Biblioteca de Genes , Ratones , Células Madre Embrionarias de Ratones , ARN no Traducido/metabolismo , Transcripción Genética
18.
Stem Cell Reports ; 14(1): 60-74, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31902705

RESUMEN

In mammals, LINE-1 (L1) retrotransposons constitute between 15% and 20% of the genome. Although only a few copies have retained the ability to retrotranspose, evidence in brain and differentiating pluripotent cells indicates that L1 retrotransposition occurs and creates mosaics in normal somatic tissues. The function of de novo insertions remains to be understood. The transdifferentiation of mouse embryonic fibroblasts to dopaminergic neuronal fate provides a suitable model for studying L1 dynamics in a defined genomic and unaltered epigenomic background. We found that L1 elements are specifically re-expressed and mobilized during the initial stages of reprogramming and that their insertions into specific acceptor loci coincides with higher chromatin accessibility and creation of new transcribed units. Those events accompany the maturation of neuronal committed cells. We conclude that L1 retrotransposition is a non-random process correlating with chromatin opening and lncRNA production that accompanies direct somatic cell reprogramming.


Asunto(s)
Transdiferenciación Celular/genética , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Elementos de Nucleótido Esparcido Largo , Animales , Biomarcadores , Técnicas de Cultivo de Célula , Línea Celular , Biología Computacional/métodos , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma , Ratones , Retroelementos , Secuenciación Completa del Genoma
19.
Epigenetics Chromatin ; 12(1): 78, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856907

RESUMEN

BACKGROUND: While the role of Polycomb group protein-mediated "cell memory" is well established in developmental contexts, little is known about their role in adult tissues and in particular in post-mitotic cells. Emerging evidence assigns a pivotal role in cell plasticity and adaptation. PRC2-Ezh1α/ß signaling pathway from cytoplasm to chromatin protects skeletal muscle cells from oxidative stress. However, detailed mechanisms controlling degradation of cytoplasmic Ezh1ß and assembly of canonical PRC2-Ezh1α repressive complex remain to be clarified. RESULTS: Here, we report NEDD4 ubiquitin E3 ligase, as key regulator of Ezh1ß. In addition, we report that ubiquitination and degradation of Ezh1ß is controlled by another layer of regulation, that is, one specific phosphorylation of serine 560 located at Ezh1ß-specific C terminal. Finally, we demonstrate that also Ezh1α needs to be stabilized under stress condition and this stabilization process requires decreased association pattern between another E3 ubiquitin ligase HUWE1. CONCLUSIONS: Together, these results shed light on key components that regulate PRC2-Ezh1α/ß pathway to direct modulation of epigenome plasticity and transcriptional output in skeletal muscle cells.


Asunto(s)
Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Estrés Oxidativo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Histonas/metabolismo , Peróxido de Hidrógeno/farmacología , Ratones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Complejo Represivo Polycomb 2/química , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Ubiquitinación
20.
Cell Syst ; 9(5): 446-458.e6, 2019 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-31629687

RESUMEN

The impact of mammalian RNA interference components, particularly, Argonaute proteins, on chromatin organization is unexplored. Recent reports indicate that AGO1 association with chromatin appears to influence gene expression. To uncover the role of AGO1 in the nucleus, we used a combination of genome-wide approaches in control and AGO1-depleted HepG2 cells. We found that AGO1 strongly associates with active enhancers and RNA being produced at those sites. Hi-C analysis revealed AGO1 enrichment at the boundaries of topologically associated domains (TADs). By Hi-C in AGO1 knockdown cells, we observed changes in chromatin organization, including TADs and A/B compartment mixing, specifically in AGO1-bound regions. Distinct groups of genes and especially eRNA transcripts located within differentially interacting loci showed altered expression upon AGO1 depletion. Moreover, AGO1 association with enhancers is dependent on eRNA transcription. Collectively, our data suggest that enhancer-associated AGO1 contributes to the fine-tuning of chromatin architecture and gene expression in human cells.


Asunto(s)
Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Factores Eucarióticos de Iniciación/genética , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica/genética , Núcleo Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Elementos de Facilitación Genéticos/genética , Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Genoma Humano/genética , Células HEK293 , Células Hep G2 , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA