Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 624(7990): 173-181, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38030723

RESUMEN

In diploid organisms, biallelic gene expression enables the production of adequate levels of mRNA1,2. This is essential for haploinsufficient genes, which require biallelic expression for optimal function to prevent the onset of developmental disorders1,3. Whether and how a biallelic or monoallelic state is determined in a cell-type-specific manner at individual loci remains unclear. MSL2 is known for dosage compensation of the male X chromosome in flies. Here we identify a role of MSL2 in regulating allelic expression in mammals. Allele-specific bulk and single-cell analyses in mouse neural progenitor cells revealed that, in addition to the targets showing biallelic downregulation, a class of genes transitions from biallelic to monoallelic expression after MSL2 loss. Many of these genes are haploinsufficient. In the absence of MSL2, one allele remains active, retaining active histone modifications and transcription factor binding, whereas the other allele is silenced, exhibiting loss of promoter-enhancer contacts and the acquisition of DNA methylation. Msl2-knockout mice show perinatal lethality and heterogeneous phenotypes during embryonic development, supporting a role for MSL2 in regulating gene dosage. The role of MSL2 in preserving biallelic expression of specific dosage-sensitive genes sets the stage for further investigation of other factors that are involved in allelic dosage compensation in mammalian cells, with considerable implications for human disease.


Asunto(s)
Alelos , Regulación de la Expresión Génica , Ubiquitina-Proteína Ligasas , Animales , Femenino , Masculino , Ratones , Metilación de ADN , Compensación de Dosificación (Genética) , Desarrollo Embrionario , Elementos de Facilitación Genéticos , Haploinsuficiencia , Histonas/metabolismo , Ratones Noqueados , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
STAR Protoc ; 4(4): 102505, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37733595

RESUMEN

We present a detailed protocol for sequencing full-length mRNA isoforms using the Oxford nanopore long-read sequencing technology. We describe steps for poly(A) RNA isolation, library preparation, and cDNA size selection. We then detail procedures for sequencing and processing and a computational framework to identify exon couplings and assign mRNA 5' ends and 3' ends to each other. Our approach enables the identification of links between transcription initiation and co-transcriptional RNA processing events. For complete details on the use and execution of this protocol, please refer to Alfonso-Gonzalez et al.1.


Asunto(s)
Procesamiento Postranscripcional del ARN , Procesamiento Postranscripcional del ARN/genética , ADN Complementario , Exones , Biblioteca de Genes , ARN Mensajero/genética
3.
Cell ; 186(11): 2438-2455.e22, 2023 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-37178687

RESUMEN

The generation of distinct messenger RNA isoforms through alternative RNA processing modulates the expression and function of genes, often in a cell-type-specific manner. Here, we assess the regulatory relationships between transcription initiation, alternative splicing, and 3' end site selection. Applying long-read sequencing to accurately represent even the longest transcripts from end to end, we quantify mRNA isoforms in Drosophila tissues, including the transcriptionally complex nervous system. We find that in Drosophila heads, as well as in human cerebral organoids, 3' end site choice is globally influenced by the site of transcription initiation (TSS). "Dominant promoters," characterized by specific epigenetic signatures including p300/CBP binding, impose a transcriptional constraint to define splice and polyadenylation variants. In vivo deletion or overexpression of dominant promoters as well as p300/CBP loss disrupted the 3' end expression landscape. Our study demonstrates the crucial impact of TSS choice on the regulation of transcript diversity and tissue identity.


Asunto(s)
Empalme Alternativo , Isoformas de ARN , Sitio de Iniciación de la Transcripción , Humanos , Poliadenilación , Regiones Promotoras Genéticas , Isoformas de ARN/metabolismo , ARN Mensajero/metabolismo
4.
J Vis Exp ; (184)2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35786676

RESUMEN

Obesity is a complex disease influenced by genetics, epigenetics, the environment, and their interactions. Mature adipocytes represent the major cell type in white adipose tissue. Understanding how adipocytes function and respond to (epi)genetic and environmental signals is essential for identifying the cause(s) of obesity. RNA and chromatin have previously been isolated from adipocytes using enzymatic digestion. In addition, protocols have been developed for nuclear isolation, where purification is achieved by fluorescence-activated cell sorting (FACS) of adipocyte-specific transgenic reporters. One of the greatest challenges to achieving high yield and quality during such protocols is the substantial amount of lipid contained in adipose tissue. The present protocol describes an optimized procedure for isolating mature adipocytes that leverages heptane to separate lipids from the targets of interest (RNA/chromatin). The resulting RNA has high integrity and generates high-quality RNA-seq results. Likewise, the procedure improves nuclei yield rate and generates reproducible ChIP-seq results across samples. Therefore, the current study provides a reliable and universal murine adipocyte isolation protocol suitable for whole-genome transcriptome and epigenome studies.


Asunto(s)
Adipocitos Blancos , Transcriptoma , Animales , Cromatina/metabolismo , Epigenoma , Ratones , Obesidad/metabolismo , ARN/metabolismo
5.
Cell Metab ; 34(4): 516-532.e11, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35316657

RESUMEN

Metabolic reprogramming is a hallmark of activated T cells. The switch from oxidative phosphorylation to aerobic glycolysis provides energy and intermediary metabolites for the biosynthesis of macromolecules to support clonal expansion and effector function. Here, we show that glycolytic reprogramming additionally controls inflammatory gene expression via epigenetic remodeling. We found that the glucose transporter GLUT3 is essential for the effector functions of Th17 cells in models of autoimmune colitis and encephalomyelitis. At the molecular level, we show that GLUT3-dependent glucose uptake controls a metabolic-transcriptional circuit that regulates the pathogenicity of Th17 cells. Metabolomic, epigenetic, and transcriptomic analyses linked GLUT3 to mitochondrial glucose oxidation and ACLY-dependent acetyl-CoA generation as a rate-limiting step in the epigenetic regulation of inflammatory gene expression. Our findings are also important from a translational perspective because inhibiting GLUT3-dependent acetyl-CoA generation is a promising metabolic checkpoint to mitigate Th17-cell-mediated inflammatory diseases.


Asunto(s)
ATP Citrato (pro-S)-Liasa , Transportador de Glucosa de Tipo 3 , Células Th17 , ATP Citrato (pro-S)-Liasa/metabolismo , Acetilcoenzima A/metabolismo , Animales , Epigénesis Genética , Glucosa/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Glucólisis/genética , Humanos , Ratones , Células Th17/metabolismo
6.
Nat Commun ; 11(1): 5200, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060580

RESUMEN

During neuronal differentiation, the transcriptional profile and the epigenetic context of neural committed cells is subject to significant rearrangements, but a systematic quantification of global histone modification changes is still missing. Here, we show that H3K79me2 increases and H3K27ac decreases globally during in-vitro neuronal differentiation of murine embryonic stem cells. DOT1L mediates all three degrees of methylation of H3K79 and its enzymatic activity is critical to modulate cellular differentiation and reprogramming. In this context, we find that inhibition of DOT1L in neural progenitor cells biases the transcriptional state towards neuronal differentiation, resulting in transcriptional upregulation of genes marked with H3K27me3 on the promoter region. We further show that DOT1L inhibition affects accessibility of SOX2-bound enhancers and impairs SOX2 binding in neural progenitors. Our work provides evidence that DOT1L activity gates differentiation of progenitors by allowing SOX2-dependent transcription of stemness programs.


Asunto(s)
Diferenciación Celular/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Proteínas Portadoras , Cromatina , Células Madre Embrionarias , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Metilación , Ratones , Células-Madre Neurales/metabolismo , Neuronas/fisiología , Procesamiento Proteico-Postraduccional/fisiología
7.
Genome Biol ; 20(1): 227, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31699133

RESUMEN

We present the software Condition-specific Regulatory Units Prediction (CRUP) to infer from epigenetic marks a list of regulatory units consisting of dynamically changing enhancers with their target genes. The workflow consists of a novel pre-trained enhancer predictor that can be reliably applied across cell types and species, solely based on histone modification ChIP-seq data. Enhancers are subsequently assigned to different conditions and correlated with gene expression to derive regulatory units. We thoroughly test and then apply CRUP to a rheumatoid arthritis model, identifying enhancer-gene pairs comprising known disease genes as well as new candidate genes.


Asunto(s)
Elementos de Facilitación Genéticos , Programas Informáticos , Animales , Artritis Experimental/genética , Artritis Reumatoide/genética , Secuenciación de Inmunoprecipitación de Cromatina , Código de Histonas , Ratones
8.
Genes Dev ; 33(21-22): 1591-1612, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31601616

RESUMEN

Genome rearrangements that occur during evolution impose major challenges on regulatory mechanisms that rely on three-dimensional genome architecture. Here, we developed a scaffolding algorithm and generated chromosome-length assemblies from Hi-C data for studying genome topology in three distantly related Drosophila species. We observe extensive genome shuffling between these species with one synteny breakpoint after approximately every six genes. A/B compartments, a set of large gene-dense topologically associating domains (TADs), and spatial contacts between high-affinity sites (HAS) located on the X chromosome are maintained over 40 million years, indicating architectural conservation at various hierarchies. Evolutionary conserved genes cluster in the vicinity of HAS, while HAS locations appear evolutionarily flexible, thus uncoupling functional requirement of dosage compensation from individual positions on the linear X chromosome. Therefore, 3D architecture is preserved even in scenarios of thousands of rearrangements highlighting its relevance for essential processes such as dosage compensation of the X chromosome.


Asunto(s)
Drosophila/genética , Evolución Molecular , Regulación de la Expresión Génica/genética , Genoma de los Insectos/genética , Algoritmos , Animales , Cromosomas de Insectos/genética , Secuencia Conservada , Compensación de Dosificación (Genética) , Drosophila/embriología , Embrión no Mamífero
9.
Commun Biol ; 1: 214, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30534606

RESUMEN

Chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) is an invaluable tool for mapping chromatin-associated proteins. Current barcoding strategies aim to improve assay throughput and scalability but intense sample handling and lack of standardization over cell types, cell numbers and epitopes hinder wide-spread use in the field. Here, we present a barcoding method to enable high-throughput ChIP-seq using common molecular biology techniques. The method, called RELACS (restriction enzyme-based labeling of chromatin in situ) relies on standardized nuclei extraction from any source and employs chromatin cutting and barcoding within intact nuclei. Barcoded nuclei are pooled and processed within the same ChIP reaction, for maximal comparability and workload reduction. The innovative barcoding concept is particularly user-friendly and suitable for implementation to standardized large-scale clinical studies and scarce samples. Aiming to maximize universality and scalability, RELACS can generate ChIP-seq libraries for transcription factors and histone modifications from hundreds of samples within three days.

10.
Hum Mol Genet ; 27(23): 4117-4134, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30452683

RESUMEN

Pluripotent stem cells are invaluable resources to study development and disease, holding a great promise for regenerative medicine. Here we use human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) from patients with Huntington's disease (HD-iPSCs) to shed light into the normal function of huntingtin (HTT) and its demise in disease. We find that HTT binds ATF7IP, a regulator of the histone H3 methyltransferase SETDB1. HTT inhibits the interaction of the ATF7IP-SETDB1 complex with other heterochromatin regulators and transcriptional repressors, maintaining low levels of H3K9 trimethylation (H3K9me3) in hESCs. Loss of HTT promotes global increased H3K9me3 levels and enrichment of H3K9me3 marks at distinct genes, including transcriptional regulators of neuronal differentiation. Although these genes are normally expressed at low amounts in hESCs, HTT knockdown (KD) reduces their induction during neural differentiation. Notably, mutant expanded polyglutamine repeats in HTT diminish its interaction with ATF7IP-SETDB1 complex and trigger H3K9me3 in HD-iPSCs. Conversely, KD of ATF7IP in HD-iPSCs reduces H3K9me3 alterations and ameliorates gene expression changes in their neural counterparts. Taken together, our results indicate ATF7IP as a potential target to correct aberrant H3K9me3 levels induced by mutant HTT.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteína Metiltransferasas/genética , Factores de Transcripción/genética , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Heterocromatina/genética , Histona Metiltransferasas/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Lentivirus/genética , Neuronas/metabolismo , Neuronas/patología , Péptidos/genética , Proteínas Represoras
11.
Genome Biol ; 19(1): 150, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30266094

RESUMEN

BACKGROUND: Partially methylated domains are extended regions in the genome exhibiting a reduced average DNA methylation level. They cover gene-poor and transcriptionally inactive regions and tend to be heterochromatic. We present a comprehensive comparative analysis of partially methylated domains in human and mouse cells, to identify structural and functional features associated with them. RESULTS: Partially methylated domains are present in up to 75% of the genome in human and mouse cells irrespective of their tissue or cell origin. Each cell type has a distinct set of partially methylated domains, and genes expressed in such domains show a strong cell type effect. The methylation level varies between cell types with a more pronounced effect in differentiating and replicating cells. The lowest level of methylation is observed in highly proliferating and immortal cancer cell lines. A decrease of DNA methylation within partially methylated domains tends to be linked to an increase in heterochromatic histone marks and a decrease of gene expression. Characteristic combinations of heterochromatic signatures in partially methylated domains are linked to domains of early and middle S-phase and late S-G2 phases of DNA replication. CONCLUSIONS: Partially methylated domains are prominent signatures of long-range epigenomic organization. Integrative analysis identifies them as important general, lineage- and cell type-specific topological features. Changes in partially methylated domains are hallmarks of cell differentiation, with decreased methylation levels and increased heterochromatic marks being linked to enhanced cell proliferation. In combination with broad histone marks, partially methylated domains demarcate distinct domains of late DNA replication.


Asunto(s)
Metilación de ADN/genética , Especificidad de Órganos/genética , Animales , Línea Celular , Replicación del ADN/genética , Genoma Humano , Heterocromatina/metabolismo , Humanos , Ratones , Neoplasias/genética , Transcripción Genética
12.
Cell Metab ; 27(6): 1294-1308.e7, 2018 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-29754954

RESUMEN

To date, it remains largely unclear to what extent chromatin machinery contributes to the susceptibility and progression of complex diseases. Here, we combine deep epigenome mapping with single-cell transcriptomics to mine for evidence of chromatin dysregulation in type 2 diabetes. We find two chromatin-state signatures that track ß cell dysfunction in mice and humans: ectopic activation of bivalent Polycomb-silenced domains and loss of expression at an epigenomically unique class of lineage-defining genes. ß cell-specific Polycomb (Eed/PRC2) loss of function in mice triggers diabetes-mimicking transcriptional signatures and highly penetrant, hyperglycemia-independent dedifferentiation, indicating that PRC2 dysregulation contributes to disease. The work provides novel resources for exploring ß cell transcriptional regulation and identifies PRC2 as necessary for long-term maintenance of ß cell identity. Importantly, the data suggest a two-hit (chromatin and hyperglycemia) model for loss of ß cell identity in diabetes.


Asunto(s)
Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Silenciador del Gen , Células Secretoras de Insulina/metabolismo , Complejo Represivo Polycomb 2/fisiología , Animales , Diferenciación Celular/genética , Células Cultivadas , Mapeo Cromosómico , Diabetes Mellitus Tipo 2/genética , Epigenómica , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Hiperglucemia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Complejo Represivo Polycomb 2/genética , Análisis de la Célula Individual
13.
Nat Commun ; 9(1): 189, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335486

RESUMEN

Despite an abundance of new studies about topologically associating domains (TADs), the role of genetic information in TAD formation is still not fully understood. Here we use our software, HiCExplorer (hicexplorer.readthedocs.io) to annotate >2800 high-resolution (570 bp) TAD boundaries in Drosophila melanogaster. We identify eight DNA motifs enriched at boundaries, including a motif bound by the M1BP protein, and two new boundary motifs. In contrast to mammals, the CTCF motif is only enriched on a small fraction of boundaries flanking inactive chromatin while most active boundaries contain the motifs bound by the M1BP or Beaf-32 proteins. We demonstrate that boundaries can be accurately predicted using only the motif sequences at open chromatin sites. We propose that DNA sequence guides the genome architecture by allocation of boundary proteins in the genome. Finally, we present an interactive online database to access and explore the spatial organization of fly, mouse and human genomes, available at http://chorogenome.ie-freiburg.mpg.de .


Asunto(s)
Cromatina/ultraestructura , Mapeo Cromosómico/métodos , Cromosomas de Insectos/ultraestructura , Drosophila melanogaster/genética , Genoma de los Insectos , Animales , Evolución Biológica , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/química , Ensamble y Desensamble de Cromatina , Cromosomas de Insectos/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Bases de Datos Genéticas , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/ultraestructura , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Expresión Génica , Humanos , Ratones , Conformación Molecular , Motivos de Nucleótidos , Programas Informáticos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Cell Death Dis ; 8(11): e3173, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29168801

RESUMEN

While several studies correlated increased expression of the histone code reader Spin1 with tumor formation or growth, little is known about physiological functions of the protein. We generated Spin1M5 mice with ablation of Spin1 in myoblast precursors using the Myf5-Cre deleter strain. Most Spin1M5 mice die shortly after birth displaying severe sarcomere disorganization and necrosis. Surviving Spin1M5 mice are growth-retarded and exhibit the most prominent defects in soleus, tibialis anterior, and diaphragm muscle. Transcriptome analyses of limb muscle at embryonic day (E) 15.5, E16.5, and at three weeks of age provided evidence for aberrant fetal myogenesis and identified deregulated skeletal muscle (SkM) functional networks. Determination of genome-wide chromatin occupancy in primary myoblast revealed direct Spin1 target genes and suggested that deregulated basic helix-loop-helix transcription factor networks account for developmental defects in Spin1M5 fetuses. Furthermore, correlating histological and transcriptome analyses, we show that aberrant expression of titin-associated proteins, abnormal glycogen metabolism, and neuromuscular junction defects contribute to SkM pathology in Spin1M5 mice. Together, we describe the first example of a histone code reader controlling SkM development in mice, which hints at Spin1 as a potential player in human SkM disease.


Asunto(s)
Proteínas de Ciclo Celular/genética , Código de Histonas/genética , Proteínas Asociadas a Microtúbulos/genética , Desarrollo de Músculos/genética , Fosfoproteínas/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Factores de Transcripción/genética
15.
Cell Death Dis ; 8(2): e2631, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28230862

RESUMEN

Coordination of energy metabolism is essential for homeostasis of stem cells, whereas an imbalance in energy homeostasis causes disease and accelerated aging. Here we show that deletion or enzymatic inactivation of lysine-specific demethylase 1 (Lsd1) triggers senescence in trophoblast stem cells (TSCs). Genome-wide transcriptional profiling of TSCs following Lsd1 inhibition shows gene set enrichment of aging and metabolic pathways. Consistently, global metabolomic and phenotypic analyses disclose an unbalanced redox status, decreased glutamine anaplerosis and mitochondrial function. Loss of homeostasis is caused by increased expression of sirtuin 4 (Sirt4), a Lsd1-repressed direct target gene. Accordingly, Sirt4 overexpression in wild-type TSCs recapitulates the senescence phenotype initiated by Lsd1 deletion or inhibition. Inversely, absence of Lsd1 enzymatic activity concomitant with knockdown of Sirt4 reestablishes normal glutamine anaplerosis, redox balance and mitochondrial function. In conclusion, by repression of Sirt4, Lsd1 directs the epigenetic control of TSC immortality via maintenance of metabolic flexibility.


Asunto(s)
Senescencia Celular/fisiología , Histona Demetilasas/metabolismo , Proteínas Mitocondriales/metabolismo , Sirtuinas/metabolismo , Células Madre/metabolismo , Trofoblastos/metabolismo , Envejecimiento/metabolismo , Humanos , Redes y Vías Metabólicas/fisiología , Mitocondrias/metabolismo , Oxidación-Reducción , Células Madre/fisiología , Trofoblastos/fisiología
16.
Immunity ; 45(5): 1148-1161, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851915

RESUMEN

The impact of epigenetics on the differentiation of memory T (Tmem) cells is poorly defined. We generated deep epigenomes comprising genome-wide profiles of DNA methylation, histone modifications, DNA accessibility, and coding and non-coding RNA expression in naive, central-, effector-, and terminally differentiated CD45RA+ CD4+ Tmem cells from blood and CD69+ Tmem cells from bone marrow (BM-Tmem). We observed a progressive and proliferation-associated global loss of DNA methylation in heterochromatic parts of the genome during Tmem cell differentiation. Furthermore, distinct gradually changing signatures in the epigenome and the transcriptome supported a linear model of memory development in circulating T cells, while tissue-resident BM-Tmem branched off with a unique epigenetic profile. Integrative analyses identified candidate master regulators of Tmem cell differentiation, including the transcription factor FOXP1. This study highlights the importance of epigenomic changes for Tmem cell biology and demonstrates the value of epigenetic data for the identification of lineage regulators.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Epigénesis Genética/inmunología , Epigenómica/métodos , Memoria Inmunológica/inmunología , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Humanos , Aprendizaje Automático , Reacción en Cadena de la Polimerasa , Transcriptoma
17.
Cell Rep ; 17(4): 1008-1021, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760309

RESUMEN

Previous work indicated that lysine-specific demethylase 1 (Lsd1) can positively regulate the oxidative and thermogenic capacities of white and beige adipocytes. Here we investigate the role of Lsd1 in brown adipose tissue (BAT) and find that BAT-selective Lsd1 ablation induces a shift from oxidative to glycolytic metabolism. This shift is associated with downregulation of BAT-specific and upregulation of white adipose tissue (WAT)-selective gene expression. This results in the accumulation of di- and triacylglycerides and culminates in a profound whitening of BAT in aged Lsd1-deficient mice. Further studies show that Lsd1 maintains BAT properties via a dual role. It activates BAT-selective gene expression in concert with the transcription factor Nrf1 and represses WAT-selective genes through recruitment of the CoREST complex. In conclusion, our data uncover Lsd1 as a key regulator of gene expression and metabolic function in BAT.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Eliminación de Gen , Histona Demetilasas/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Regulación de la Expresión Génica , Glucosa/metabolismo , Glucólisis/genética , Metabolismo de los Lípidos/genética , Ratones Noqueados , Modelos Biológicos , Oxidación-Reducción , Aumento de Peso
18.
Artículo en Inglés | MEDLINE | ID: mdl-27478504

RESUMEN

BACKGROUND: Monocyte-to-macrophage differentiation involves major biochemical and structural changes. In order to elucidate the role of gene regulatory changes during this process, we used high-throughput sequencing to analyze the complete transcriptome and epigenome of human monocytes that were differentiated in vitro by addition of colony-stimulating factor 1 in serum-free medium. RESULTS: Numerous mRNAs and miRNAs were significantly up- or down-regulated. More than 100 discrete DNA regions, most often far away from transcription start sites, were rapidly demethylated by the ten eleven translocation enzymes, became nucleosome-free and gained histone marks indicative of active enhancers. These regions were unique for macrophages and associated with genes involved in the regulation of the actin cytoskeleton, phagocytosis and innate immune response. CONCLUSIONS: In summary, we have discovered a phagocytic gene network that is repressed by DNA methylation in monocytes and rapidly de-repressed after the onset of macrophage differentiation.

19.
Biochim Biophys Acta ; 1859(2): 315-23, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26619801

RESUMEN

BACKGROUND: It is still unclear whether oxidative stress (OS) is a disease consequence or is directly involved in the etiology of neurodegenerative disorders (NDs) onset and/or progression; however, many of these conditions are associated with increased levels of oxidation markers and damaged cell components. Previously we demonstrated the accumulation of reactive oxygen species (ROS) and increased SOD1 gene expression in H2O2-treated SH-SY5Y cells, recapitulating pathological features of Amyotrophic Lateral Sclerosis (ALS). Since we observed a post-transcriptional regulation of SOD1 gene in this cellular model, we investigated the transcriptional regulation of SOD1 mRNA under oxidative stress (OS). RESULTS: In response to H2O2 treatment, PolII increased its association to SOD1 promoter. Electrophoretic mobility shift assays and mass spectrometry analyses on SOD1 promoter highlighted the formation of a transcriptional complex bound to the ARE sequences. Western Blotting experiments showed that in our in vitro model, H2O2 exposure increases Nrf2 expression in the nuclear fraction while immunoprecipitation confirmed its phosphorylation and release from Keap1 inhibition. However, H2O2 treatment did not modify Nrf2 binding on SOD1 promoter, which seems to be regulated by different transcription factors (TFs). CONCLUSIONS: Although our data suggest that SOD1 is transcriptionally regulated in response to OS, Nrf2 does not appear to associate with SOD1 promoter in this cellular model of neurodegeneration. Our results open new perspectives in the comprehension of two key antioxidant pathways involved in neurodegenerative disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Factor 2 Relacionado con NF-E2/biosíntesis , Degeneración Nerviosa/genética , Superóxido Dismutasa/biosíntesis , Transcripción Genética , Esclerosis Amiotrófica Lateral/patología , Línea Celular , Regulación de la Expresión Génica/genética , Humanos , Peróxido de Hidrógeno/toxicidad , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Proteína 1 Asociada A ECH Tipo Kelch , Factor 2 Relacionado con NF-E2/genética , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Estrés Oxidativo/genética , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
20.
Nucleic Acids Res ; 44(7): e67, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26704968

RESUMEN

Chromatin immunoprecipitation followed by next generation sequencing (ChIP-seq) is a key technique in chromatin research. Although heavily applied, existing ChIP-seq protocols are often highly fine-tuned workflows, optimized for specific experimental requirements. Especially the initial steps of ChIP-seq, particularly chromatin shearing, are deemed to be exceedingly cell-type-specific, thus impeding any protocol standardization efforts. Here we demonstrate that harmonization of ChIP-seq workflows across cell types and conditions is possible when obtaining chromatin from properly isolated nuclei. We established an ultrasound-based nuclei extraction method (NEXSON: Nuclei EXtraction by SONication) that is highly effective across various organisms, cell types and cell numbers. The described method has the potential to replace complex cell-type-specific, but largely ineffective, nuclei isolation protocols. By including NEXSON in ChIP-seq workflows, we completely eliminate the need for extensive optimization and sample-dependent adjustments. Apart from this significant simplification, our approach also provides the basis for a fully standardized ChIP-seq and yields highly reproducible transcription factor and histone modifications maps for a wide range of different cell types. Even small cell numbers (∼10,000 cells per ChIP) can be easily processed without application of modified chromatin or library preparation protocols.


Asunto(s)
Fraccionamiento Celular/normas , Inmunoprecipitación de Cromatina/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Animales , Fraccionamiento Celular/métodos , Línea Celular , Núcleo Celular/genética , Células Cultivadas , Cromatina/aislamiento & purificación , Femenino , Células Hep G2 , Histonas/metabolismo , Humanos , Masculino , Ratones , Reproducibilidad de los Resultados , Sonicación , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...