Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Nat Metab ; 6(6): 1053-1075, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38684889

RESUMEN

Promoting brown adipose tissue (BAT) activity innovatively targets obesity and metabolic disease. While thermogenic activation of BAT is well understood, the rheostatic regulation of BAT to avoid excessive energy dissipation remains ill-defined. Here, we demonstrate that adenylyl cyclase 3 (AC3) is key for BAT function. We identified a cold-inducible promoter that generates a 5' truncated AC3 mRNA isoform (Adcy3-at), whose expression is driven by a cold-induced, truncated isoform of PPARGC1A (PPARGC1A-AT). Male mice lacking Adcy3-at display increased energy expenditure and are resistant to obesity and ensuing metabolic imbalances. Mouse and human AC3-AT are retained in the endoplasmic reticulum, unable to translocate to the plasma membrane and lack enzymatic activity. AC3-AT interacts with AC3 and sequesters it in the endoplasmic reticulum, reducing the pool of adenylyl cyclases available for G-protein-mediated cAMP synthesis. Thus, AC3-AT acts as a cold-induced rheostat in BAT, limiting adverse consequences of cAMP activity during chronic BAT activation.


Asunto(s)
Adenilil Ciclasas , Tejido Adiposo Pardo , Frío , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Tejido Adiposo Pardo/metabolismo , Animales , Ratones , Masculino , Humanos , Termogénesis/genética , Metabolismo Energético , AMP Cíclico/metabolismo , Ratones Noqueados
2.
Hum Mol Genet ; 32(7): 1083-1089, 2023 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-36300302

RESUMEN

Auditory synaptopathy/neuropathy (AS/AN) is a distinct type of sensorineural hearing loss in which the cochlear sensitivity to sound (i.e. active cochlear amplification by outer hair cells) is preserved whereas sound encoding by inner hair cells and/or auditory nerve fibers is disrupted owing to genetic or environmental factors. Autosomal-dominant auditory neuropathy type 2 (AUNA2) was linked either to chromosomal bands 12q24 or 13q34 in a large German family in 2017. By whole-genome sequencing, we now detected a 5500 bp deletion in ATP11A on chromosome 13q34 segregating with the phenotype in this family. ATP11A encodes a P-type ATPase that translocates phospholipids from the exoplasmic to the cytoplasmic leaflet of the plasma membrane. The deletion affects both isoforms of ATP11A and activates a cryptic splice site leading to the formation of an alternative last exon. ATP11A carrying the altered C-terminus loses its flippase activity for phosphatidylserine. Atp11a is expressed in fibers and synaptic contacts of the auditory nerve and in the cochlear nucleus in mice, and conditional Atp11a knockout mice show a progressive reduction of the spiral ganglion neuron compound action potential, recapitulating the human phenotype of AN. By combining whole-genome sequencing, immunohistochemistry, in vitro functional assays and generation of a mouse model, we could thus identify a partial deletion of ATP11A as the genetic cause of AUNA2.


Asunto(s)
Pérdida Auditiva Central , Pérdida Auditiva Sensorineural , Humanos , Ratones , Animales , Pérdida Auditiva Central/genética , Pérdida Auditiva Sensorineural/genética , Mutación , Células Ciliadas Auditivas Internas , Cromosomas , Transportadoras de Casetes de Unión a ATP/genética
3.
PLoS Pathog ; 17(9): e1009943, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34555129

RESUMEN

Regulation of cellular metabolism is now recognized as a crucial mechanism for the activation of innate and adaptive immune cells upon diverse extracellular stimuli. Macrophages, for instance, increase glycolysis upon stimulation with pathogen-associated molecular patterns (PAMPs). Conceivably, pathogens also counteract these metabolic changes for their own survival in the host. Despite this dynamic interplay in host-pathogen interactions, the role of immunometabolism in the context of intracellular bacterial infections is still unclear. Here, employing unbiased metabolomic and transcriptomic approaches, we investigated the role of metabolic adaptations of macrophages upon Salmonella enterica serovar Typhimurium (S. Typhimurium) infections. Importantly, our results suggest that S. Typhimurium abrogates glycolysis and its modulators such as insulin-signaling to impair macrophage defense. Mechanistically, glycolysis facilitates glycolytic enzyme aldolase A mediated v-ATPase assembly and the acidification of phagosomes which is critical for lysosomal degradation. Thus, impairment in the glycolytic machinery eventually leads to decreased bacterial clearance and antigen presentation in murine macrophages (BMDM). Collectively, our results highlight a vital molecular link between metabolic adaptation and phagosome maturation in macrophages, which is targeted by S. Typhimurium to evade cell-autonomous defense.


Asunto(s)
Glucólisis/fisiología , Interacciones Huésped-Patógeno/fisiología , Macrófagos/metabolismo , Fagosomas/metabolismo , Salmonelosis Animal/metabolismo , Animales , Perfilación de la Expresión Génica , Metabolómica , Ratones , Salmonella typhimurium/metabolismo
4.
Nat Commun ; 11(1): 644, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005828

RESUMEN

Obesity and type 2 diabetes mellitus are global emergencies and long noncoding RNAs (lncRNAs) are regulatory transcripts with elusive functions in metabolism. Here we show that a high fraction of lncRNAs, but not protein-coding mRNAs, are repressed during diet-induced obesity (DIO) and refeeding, whilst nutrient deprivation induced lncRNAs in mouse liver. Similarly, lncRNAs are lost in diabetic humans. LncRNA promoter analyses, global cistrome and gain-of-function analyses confirm that increased MAFG signaling during DIO curbs lncRNA expression. Silencing Mafg in mouse hepatocytes and obese mice elicits a fasting-like gene expression profile, improves glucose metabolism, de-represses lncRNAs and impairs mammalian target of rapamycin (mTOR) activation. We find that obesity-repressed LincIRS2 is controlled by MAFG and observe that genetic and RNAi-mediated LincIRS2 loss causes elevated blood glucose, insulin resistance and aberrant glucose output in lean mice. Taken together, we identify a MAFG-lncRNA axis controlling hepatic glucose metabolism in health and metabolic disease.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Glucosa/metabolismo , Hígado/metabolismo , Factor de Transcripción MafG/genética , Obesidad/genética , ARN Largo no Codificante/genética , Proteínas Represoras/genética , Anciano , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Factor de Transcripción MafG/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Obesidad/metabolismo , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
5.
Sci Rep ; 9(1): 11071, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31363146

RESUMEN

AATF is a central regulator of the cellular outcome upon p53 activation, a finding that has primarily been attributed to its function as a transcription factor. Recent data showed that AATF is essential for ribosome biogenesis and plays a role in rRNA maturation. AATF has been implicated to fulfil this role through direct interaction with rRNA and was identified in several RNA-interactome capture experiments. Here, we provide a first comprehensive analysis of the RNA bound by AATF using CLIP-sequencing. Interestingly, this approach shows predominant binding of the 45S pre-ribosomal RNA precursor molecules. Furthermore, AATF binds to mRNAs encoding for ribosome biogenesis factors as well as snoRNAs. These findings are complemented by an in-depth analysis of the protein interactome of AATF containing a large set of proteins known to play a role in rRNA maturation with an emphasis on the protein-RNA-complexes known to be required for the generation of the small ribosomal subunit (SSU). In line with this finding, the binding sites of AATF within the 45S rRNA precursor localize in close proximity to the SSU cleavage sites. Consequently, our multilayer analysis of the protein-RNA interactome of AATF reveals this protein to be an important hub for protein and RNA interactions involved in ribosome biogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Represoras/metabolismo , Proteínas Ribosómicas/metabolismo , Subunidades Ribosómicas Pequeñas/metabolismo , Ribosomas/metabolismo , Animales , Sitios de Unión , Línea Celular , Células HEK293 , Humanos , Ratones , Unión Proteica , Precursores del ARN/metabolismo
6.
Nat Commun ; 10(1): 3323, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31346165

RESUMEN

Most manipulations that extend lifespan also increase resistance to various stress factors and environmental cues in a range of animals from yeast to mammals. However, the underlying molecular mechanisms regulating stress resistance during aging are still largely unknown. Here we identify Krüppel-like factor 1 (KLF-1) as a mediator of a cytoprotective response that dictates longevity induced by reduced mitochondrial function. A redox-regulated KLF-1 activation and transfer to the nucleus coincides with the peak of somatic mitochondrial biogenesis that occurs around a transition from larval stage L3 to D1. We further show that KLF-1 activates genes involved in the xenobiotic detoxification programme and identified cytochrome P450 oxidases, the KLF-1 main effectors, as longevity-assurance factors of mitochondrial mutants. Collectively, these findings underline the importance of the xenobiotic detoxification in the mitohormetic, longevity assurance pathway and identify KLF-1 as a central factor in orchestrating this response.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Mitocondrias/genética , Xenobióticos/metabolismo , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Femenino , Inactivación Metabólica , Factores de Transcripción de Tipo Kruppel/genética , Longevidad , Masculino , Mitocondrias/metabolismo
7.
Cell Stem Cell ; 24(5): 736-752.e12, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30982769

RESUMEN

The pathological consequences of structural variants disrupting 3D genome organization can be difficult to elucidate in vivo due to differences in gene dosage sensitivity between mice and humans. This is illustrated by branchiooculofacial syndrome (BOFS), a rare congenital disorder caused by heterozygous mutations within TFAP2A, a neural crest regulator for which humans, but not mice, are haploinsufficient. Here, we present a BOFS patient carrying a heterozygous inversion with one breakpoint located within a topologically associating domain (TAD) containing enhancers essential for TFAP2A expression in human neural crest cells (hNCCs). Using patient-specific hiPSCs, we show that, although the inversion shuffles the TFAP2A hNCC enhancers with novel genes within the same TAD, this does not result in enhancer adoption. Instead, the inversion disconnects one TFAP2A allele from its cognate enhancers, leading to monoallelic and haploinsufficient TFAP2A expression in patient hNCCs. Our work illustrates the power of hiPSC differentiation to unveil long-range pathomechanisms.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Variación Estructural del Genoma/genética , Mutación/genética , Cresta Neural/fisiología , Factor de Transcripción AP-2/metabolismo , Adolescente , Alelos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Elementos de Facilitación Genéticos/genética , Haploinsuficiencia , Humanos , Masculino , Ratones , Análisis de la Célula Individual , Factor de Transcripción AP-2/genética
8.
BMC Genomics ; 20(1): 85, 2019 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-30678634

RESUMEN

BACKGROUND: Next-Generation Sequencing (NGS) has been widely accepted as an essential tool in molecular biology. Reduced costs and automated analysis pipelines make the use of NGS data feasible even for small labs, yet the methods for interpreting the data are not sophisticated enough to account for the amount of information. RESULTS: We propose s ·nr, a Visual Analytics tool that provides simple yet powerful visual interfaces for displaying and querying NGS data. It allows researchers to explore their own data in the context of experimental data deposited in public repositories, as well as to extract specific data sets with similar gene expression signatures. We tested s ·nr on 1543 RNA-Seq based mouse differential expression profiles derived from the public ArrayExpress platform. We provide the repository of processed data with this paper. CONCLUSION: s ·nr, easily deployable utilizing its containerized implementation, empowers researchers to analyze and relate their own RNA-Seq as well as to provide interactive and contextual crosstalk with data from public repositories. This allows users to deduce novel and unbiased hypotheses about the underlying molecular processes. DEMO: Login demo/demo: snr.sf.mpg.de (Tested with Google Chrome).


Asunto(s)
Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Animales , Gráficos por Computador , Ratones , Interfaz Usuario-Computador
9.
Cells ; 8(1)2019 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-30669443

RESUMEN

Autophagy is a highly conserved intracellular degradative pathway that is crucial for cellular homeostasis. During autophagy, the core autophagy protein ATG12 plays, together with ATG5 and ATG16, an essential role in the expansion of the autophagosomal membrane. In this study we analyzed gene replacement mutants of atg12 in Dictyostelium discoideum AX2 wild-type and ATG16‾ cells. RNAseq analysis revealed a strong enrichment of, firstly, autophagy genes among the up-regulated genes and, secondly, genes implicated in cell motility and phagocytosis among the down-regulated genes in the generated ATG12‾, ATG16‾ and ATG12‾/16‾ cells. The mutant strains showed similar defects in fruiting body formation, autolysosome maturation, and cellular viability, implying that ATG12 and ATG16 act as a functional unit in canonical autophagy. In contrast, ablation of ATG16 or of ATG12 and ATG16 resulted in slightly more severe defects in axenic growth, macropinocytosis, and protein homeostasis than ablation of only ATG12, suggesting that ATG16 fulfils an additional function in these processes. Phagocytosis of yeast, spore viability, and maximal cell density were much more affected in ATG12‾/16‾ cells, indicating that both proteins also have cellular functions independent of each other. In summary, we show that ATG12 and ATG16 fulfil autophagy-independent functions in addition to their role in canonical autophagy.


Asunto(s)
Proteína 12 Relacionada con la Autofagia/metabolismo , Autofagia , Dictyostelium/metabolismo , Secuencia de Aminoácidos , Autofagia/genética , Proteína 12 Relacionada con la Autofagia/química , Supervivencia Celular , Secuencia Conservada , Endocitosis , Evolución Molecular , Regulación de la Expresión Génica , Ontología de Genes , Lisosomas/metabolismo , Mutación/genética , Nitrógeno/deficiencia , Proteostasis
11.
Nat Commun ; 9(1): 3622, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30190464

RESUMEN

Increasing brown adipose tissue (BAT) thermogenesis in mice and humans improves metabolic health and understanding BAT function is of interest for novel approaches to counteract obesity. The role of long noncoding RNAs (lncRNAs) in these processes remains elusive. We observed maternally expressed, imprinted lncRNA H19 increased upon cold-activation and decreased in obesity in BAT. Inverse correlations of H19 with BMI were also observed in humans. H19 overexpression promoted, while silencing of H19 impaired adipogenesis, oxidative metabolism and mitochondrial respiration in brown but not white adipocytes. In vivo, H19 overexpression protected against DIO, improved insulin sensitivity and mitochondrial biogenesis, whereas fat H19 loss sensitized towards HFD weight gains. Strikingly, paternally expressed genes (PEG) were largely absent from BAT and we demonstrated that H19 recruits PEG-inactivating H19-MBD1 complexes and acts as BAT-selective PEG gatekeeper. This has implications for our understanding how monoallelic gene expression affects metabolism in rodents and, potentially, humans.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Impresión Genómica , Obesidad/genética , ARN Largo no Codificante/genética , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Obesidad/etiología
12.
BMC Bioinformatics ; 19(1): 156, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29699486

RESUMEN

BACKGROUND: Recent cancer genome studies on many human cancer types have relied on multiple molecular high-throughput technologies. Given the vast amount of data that has been generated, there are surprisingly few databases which facilitate access to these data and make them available for flexible analysis queries in the broad research community. If used in their entirety and provided at a high structural level, these data can be directed into constantly increasing databases which bear an enormous potential to serve as a basis for machine learning technologies with the goal to support research and healthcare with predictions of clinically relevant traits. RESULTS: We have developed the Cancer Systems Biology Database (CancerSysDB), a resource for highly flexible queries and analysis of cancer-related data across multiple data types and multiple studies. The CancerSysDB can be adopted by any center for the organization of their locally acquired data and its integration with publicly available data from multiple studies. A publicly available main instance of the CancerSysDB can be used to obtain highly flexible queries across multiple data types as shown by highly relevant use cases. In addition, we demonstrate how the CancerSysDB can be used for predictive cancer classification based on whole-exome data from 9091 patients in The Cancer Genome Atlas (TCGA) research network. CONCLUSIONS: Our database bears the potential to be used for large-scale integrative queries and predictive analytics of clinically relevant traits.


Asunto(s)
Bases de Datos Factuales , Genómica/métodos , Aprendizaje Automático , Neoplasias/genética , Programas Informáticos , Biología de Sistemas , Exoma , Humanos
13.
Oncogene ; 37(11): 1503-1518, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29321668

RESUMEN

A fundamental principle in malignant tranformation is the ability of cancer cells to escape the naturally occurring cell-intrinsic responses to DNA damage. Tumors progress despite the accumulation of DNA lesions. However, the underlying mechanisms of this tolerance to genotoxic stress are still poorly characterized. Here, we show that replication stress occurs in Kras-driven murine lung adenocarcinomas, as well as in proliferating murine embryonic and adult tissues. We identify the transcriptional regulator AATF/CHE-1 as a key molecule to sustain proliferative tissues and tumor progression in parts by inhibiting p53-driven apoptosis in vivo. In an autochthonous Kras-driven lung adenocarcinoma model, deletion of Aatf delayed lung cancer formation predominantly in a p53-dependent manner. Moreover, targeting Aatf in existing tumors through a dual recombinase strategy caused a halt in tumor progression. Taken together, these data suggest that AATF may serve as a drug target to treat KRAS-driven malignancies.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/genética , Proliferación Celular/genética , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Represoras/fisiología , Adenocarcinoma del Pulmón/patología , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Embrión de Mamíferos , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína p53 Supresora de Tumor/fisiología
14.
Comput Struct Biotechnol J ; 15: 471-477, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29158876

RESUMEN

While Next-Generation Sequencing (NGS) can now be considered an established analysis technology for research applications across the life sciences, the analysis workflows still require substantial bioinformatics expertise. Typical challenges include the appropriate selection of analytical software tools, the speedup of the overall procedure using HPC parallelization and acceleration technology, the development of automation strategies, data storage solutions and finally the development of methods for full exploitation of the analysis results across multiple experimental conditions. Recently, NGS has begun to expand into clinical environments, where it facilitates diagnostics enabling personalized therapeutic approaches, but is also accompanied by new technological, legal and ethical challenges. There are probably as many overall concepts for the analysis of the data as there are academic research institutions. Among these concepts are, for instance, complex IT architectures developed in-house, ready-to-use technologies installed on-site as well as comprehensive Everything as a Service (XaaS) solutions. In this mini-review, we summarize the key points to consider in the setup of the analysis architectures, mostly for scientific rather than diagnostic purposes, and provide an overview of the current state of the art and challenges of the field.

15.
Hum Mutat ; 38(10): 1325-1335, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28598576

RESUMEN

Next-generation sequencing (NGS) has turned from a new and experimental technology into a standard procedure for cancer genome studies and clinical investigation. While a multitude of software packages for cancer genome data analysis have been made available, these need to be combined into efficient analytical workflows that cover multiple aspects relevant to a clinical environment and that deliver handy results within a reasonable time frame. Here, we introduce QuickNGS Cancer as a new suite of bioinformatics pipelines that is focused on cancer genomics and significantly reduces the analytical hurdles that still limit a broader applicability of NGS technology, particularly to clinically driven research. QuickNGS Cancer allows a highly efficient analysis of a broad variety of NGS data types, specifically considering cancer-specific issues, such as biases introduced by tumor impurity and aneuploidy or the assessment of genomic variations regarding their biomedical relevance. It delivers highly reproducible analysis results ready for interpretation within only a few days after sequencing, as shown by a reanalysis of 140 tumor/normal pairs from The Cancer Genome Atlas (TCGA) in which QuickNGS Cancer detected a significant number of mutations in key cancer genes missed by a well-established mutation calling pipeline. Finally, QuickNGS Cancer obtained several unexpected mutations in leukemias that could be confirmed by Sanger sequencing.


Asunto(s)
Genoma Humano/genética , Mutación/genética , Neoplasias/genética , Programas Informáticos , Biología Computacional , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN , Flujo de Trabajo
16.
Elife ; 62017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28632132

RESUMEN

In the arcuate nucleus of the hypothalamus (ARH) satiety signaling (anorexigenic) pro-opiomelanocortin (POMC)-expressing and hunger signaling (orexigenic) agouti-related peptide (AgRP)-expressing neurons are key components of the neuronal circuits that control food intake and energy homeostasis. Here, we assessed whether the catecholamine noradrenalin directly modulates the activity of these neurons in mice. Perforated patch clamp recordings showed that noradrenalin changes the activity of these functionally antagonistic neurons in opposite ways, increasing the activity of the orexigenic NPY/AgRP neurons and decreasing the activity of the anorexigenic POMC neurons. Cell type-specific transcriptomics and pharmacological experiments revealed that the opposing effect on these neurons is mediated by the activation of excitatory α1A - and ß- adrenergic receptors in NPY/AgRP neurons, while POMC neurons are inhibited via α2A - adrenergic receptors. Thus, the coordinated differential modulation of the key hypothalamic neurons in control of energy homeostasis assigns noradrenalin an important role to promote feeding.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/metabolismo , Proopiomelanocortina/metabolismo , Animales , Perfilación de la Expresión Génica , Ratones , Técnicas de Placa-Clamp
17.
Cell Stem Cell ; 20(5): 689-705.e9, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28285903

RESUMEN

Poised enhancers marked by H3K27me3 in pluripotent stem cells have been implicated in the establishment of somatic expression programs during embryonic stem cell (ESC) differentiation. However, the functional relevance and mechanism of action of poised enhancers remain unknown. Using CRISPR/Cas9 technology to engineer precise genetic deletions, we demonstrate that poised enhancers are necessary for the induction of major anterior neural regulators. Interestingly, circularized chromosome conformation capture sequencing (4C-seq) shows that poised enhancers already establish physical interactions with their target genes in ESCs in a polycomb repressive complex 2 (PRC2)-dependent manner. Loss of PRC2 does not activate poised enhancers or induce their putative target genes in undifferentiated ESCs; however, loss of PRC2 in differentiating ESCs severely and specifically compromises the induction of major anterior neural genes representing poised enhancer targets. Overall, our work illuminates an unexpected function for polycomb proteins in facilitating neural induction by endowing major anterior neural loci with a permissive regulatory topology.


Asunto(s)
Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Ratones , Complejo Represivo Polycomb 2/genética , Reacción en Cadena de la Polimerasa
18.
Cell Rep ; 17(11): 3062-3076, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27974217

RESUMEN

Cellular heterogeneity within embryonic and adult tissues is involved in multiple biological and pathological processes. Here, we present a simple epigenomic strategy that allows the functional dissection of cellular heterogeneity. By integrating H3K27me3 chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq) data, we demonstrate that the presence of broad H3K27me3 domains at transcriptionally active genes reflects the heterogeneous expression of major cell identity regulators. Using dorsoventral patterning of the spinal neural tube as a model, the proposed approach successfully identifies the majority of previously known dorsoventral patterning transcription factors with high sensitivity and precision. Moreover, poorly characterized patterning regulators can be similarly predicted, as shown for ZNF488, which confers p1/p2 neural progenitor identity. Finally, we show that, as our strategy is based on universal chromatin features, it can be used to functionally dissect cellular heterogeneity within various organisms and tissues, thus illustrating its potential applicability to a broad range of biological and pathological contexts.


Asunto(s)
Tipificación del Cuerpo/genética , Linaje de la Célula/genética , Epigenómica , Heterogeneidad Genética , Animales , Cromatina/genética , Inmunoprecipitación de Cromatina/métodos , Regulación del Desarrollo de la Expresión Génica , Genoma , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Tubo Neural/crecimiento & desarrollo , Tubo Neural/metabolismo , Análisis de Secuencia de ARN/métodos , Columna Vertebral/crecimiento & desarrollo , Columna Vertebral/metabolismo
19.
BMC Genomics ; 17(1): 855, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27809788

RESUMEN

BACKGROUND: Although the pattern of lateral organ formation from apical meristems establishes species-specific plant architecture, the positional information that confers cell fate to cells as they transit to the meristem flanks where they differentiate, remains largely unknown. We have combined fluorescence-activated cell sorting and RNA-seq to characterise the cell-type-specific transcriptome at the earliest developmental time-point of lateral organ formation using DORNRÖSCHEN-LIKE::GFP to mark founder-cell populations at the periphery of the inflorescence meristem (IM) in apetala1 cauliflower double mutants, which overproliferate IMs. RESULTS: Within the lateral organ founder-cell population at the inflorescence meristem, floral primordium identity genes are upregulated and stem-cell identity markers are downregulated. Additional differentially expressed transcripts are involved in polarity generation and boundary formation, and in epigenetic and post-translational changes. However, only subtle transcriptional reprogramming within the global auxin network was observed. CONCLUSIONS: The transcriptional network of differentially expressed genes supports the hypothesis that lateral organ founder-cell specification involves the creation of polarity from the centre to the periphery of the IM and the establishment of a boundary from surrounding cells, consistent with bract initiation. However, contrary to the established paradigm that sites of auxin response maxima pre-pattern lateral organ initiation in the IM, auxin response might play a minor role in the earliest stages of lateral floral initiation.


Asunto(s)
Brassica/genética , Inflorescencia/genética , Meristema/genética , Transcriptoma , Análisis por Conglomerados , Biología Computacional/métodos , Epigénesis Genética , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Ontología de Genes , Redes Reguladoras de Genes , Genes Reporteros , Fenotipo , Células Vegetales/metabolismo , Procesamiento Postranscripcional del ARN
20.
Cell ; 165(1): 125-138, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27015310

RESUMEN

Activation of Agouti-related peptide (AgRP) neurons potently promotes feeding, and chronically altering their activity also affects peripheral glucose homeostasis. We demonstrate that acute activation of AgRP neurons causes insulin resistance through impairment of insulin-stimulated glucose uptake into brown adipose tissue (BAT). AgRP neuron activation acutely reprograms gene expression in BAT toward a myogenic signature, including increased expression of myostatin. Interference with myostatin activity improves insulin sensitivity that was impaired by AgRP neurons activation. Optogenetic circuitry mapping reveals that feeding and insulin sensitivity are controlled by both distinct and overlapping projections. Stimulation of AgRP → LHA projections impairs insulin sensitivity and promotes feeding while activation of AgRP → anterior bed nucleus of the stria terminalis (aBNST)vl projections, distinct from AgRP → aBNSTdm projections controlling feeding, mediate the effect of AgRP neuron activation on BAT-myostatin expression and insulin sensitivity. Collectively, our results suggest that AgRP neurons in mice induce not only eating, but also insulin resistance by stimulating expression of muscle-related genes in BAT, revealing a mechanism by which these neurons rapidly coordinate hunger states with glucose homeostasis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Regulación del Apetito , Glucosa/metabolismo , Resistencia a la Insulina , Neuronas/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Conducta Alimentaria , Ratones , Miostatina/genética , Optogenética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...