Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(13): 3338-3356.e30, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38810644

RESUMEN

Suspended animation states allow organisms to survive extreme environments. The African turquoise killifish has evolved diapause as a form of suspended development to survive a complete drought. However, the mechanisms underlying the evolution of extreme survival states are unknown. To understand diapause evolution, we performed integrative multi-omics (gene expression, chromatin accessibility, and lipidomics) in the embryos of multiple killifish species. We find that diapause evolved by a recent remodeling of regulatory elements at very ancient gene duplicates (paralogs) present in all vertebrates. CRISPR-Cas9-based perturbations identify the transcription factors REST/NRSF and FOXOs as critical for the diapause gene expression program, including genes involved in lipid metabolism. Indeed, diapause shows a distinct lipid profile, with an increase in triglycerides with very-long-chain fatty acids. Our work suggests a mechanism for the evolution of complex adaptations and offers strategies to promote long-term survival by activating suspended animation programs in other species.


Asunto(s)
Diapausa , Animales , Evolución Biológica , Diapausa/genética , Embrión no Mamífero/metabolismo , Fundulidae/genética , Fundulidae/metabolismo , Regulación del Desarrollo de la Expresión Génica , Peces Killi/genética , Peces Killi/metabolismo , Metabolismo de los Lípidos/genética , Proteínas de Peces/genética , Masculino , Femenino
2.
Nat Rev Mol Cell Biol ; 24(1): 45-62, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35859206

RESUMEN

Most adult organs contain regenerative stem cells, often organized in specific niches. Stem cell function is critical for tissue homeostasis and repair upon injury, and it is dependent on interactions with the niche. During ageing, stem cells decline in their regenerative potential and ability to give rise to differentiated cells in the tissue, which is associated with a deterioration of tissue integrity and health. Ageing-associated changes in regenerative tissue regions include defects in maintenance of stem cell quiescence, differentiation ability and bias, clonal expansion and infiltration of immune cells in the niche. In this Review, we discuss cellular and molecular mechanisms underlying ageing in the regenerative regions of different tissues as well as potential rejuvenation strategies. We focus primarily on brain, muscle and blood tissues, but also provide examples from other tissues, such as skin and intestine. We describe the complex interactions between different cell types, non-cell-autonomous mechanisms between ageing niches and stem cells, and the influence of systemic factors. We also compare different interventions for the rejuvenation of old regenerative regions. Future outlooks in the field of stem cell ageing are discussed, including strategies to counter ageing and age-dependent disease.


Asunto(s)
Rejuvenecimiento , Células Madre , Senescencia Celular/fisiología , Diferenciación Celular , Nicho de Células Madre
3.
Cell ; 177(1): 200-220, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30901541

RESUMEN

Aging negatively impacts vitality and health. Many genetic pathways that regulate aging were discovered in invertebrates. However, the genetics of aging is more complex in vertebrates because of their specialized systems. This Review discusses advances in the genetic regulation of aging in vertebrates from work in mice, humans, and organisms with exceptional lifespans. We highlight challenges for the future, including sex-dependent differences in lifespan and the interplay between genes and environment. We also discuss how the identification of reliable biomarkers of age and development of new vertebrate models can be leveraged for personalized interventions to counter aging and age-related diseases.


Asunto(s)
Envejecimiento/genética , Longevidad/genética , Vertebrados/genética , Animales , Envejecimiento Saludable/genética , Humanos , Invertebrados/genética , Ratones , Modelos Animales
4.
Cell ; 167(7): 1672-1674, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984716

RESUMEN

In a thought-provoking study, Ocampo et al. show that the cyclic expression of stem cell reprogramming factors in vivo increases the lifespan of a mouse model of premature aging and provides health benefits to chronologically old, normal mice.


Asunto(s)
Envejecimiento/metabolismo , Reprogramación Celular , Animales , Ratones
5.
Cell ; 160(5): 1013-1026, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25684364

RESUMEN

VIDEO ABSTRACT: Aging is a complex process that affects multiple organs. Modeling aging and age-related diseases in the lab is challenging because classical vertebrate models have relatively long lifespans. Here, we develop the first platform for rapid exploration of age-dependent traits and diseases in vertebrates, using the naturally short-lived African turquoise killifish. We provide an integrative genomic and genome-editing toolkit in this organism using our de-novo-assembled genome and the CRISPR/Cas9 technology. We mutate many genes encompassing the hallmarks of aging, and for a subset, we produce stable lines within 2-3 months. As a proof of principle, we show that fish deficient for the protein subunit of telomerase exhibit the fastest onset of telomere-related pathologies among vertebrates. We further demonstrate the feasibility of creating specific genetic variants. This genome-to-phenotype platform represents a unique resource for studying vertebrate aging and disease in a high-throughput manner and for investigating candidates arising from human genome-wide studies.


Asunto(s)
Peces Killi/fisiología , Envejecimiento , Animales , Secuencia de Bases , Sistemas CRISPR-Cas , ADN Polimerasa Dirigida por ADN/metabolismo , Femenino , Técnicas Genéticas , Humanos , Peces Killi/genética , Masculino , Modelos Animales , Datos de Secuencia Molecular , Telomerasa/genética , Telomerasa/metabolismo , Vertebrados/fisiología
6.
Cell ; 163(6): 1539-54, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26638078

RESUMEN

Lifespan is a remarkably diverse trait ranging from a few days to several hundred years in nature, but the mechanisms underlying the evolution of lifespan differences remain elusive. Here we de novo assemble a reference genome for the naturally short-lived African turquoise killifish, providing a unique resource for comparative and experimental genomics. The identification of genes under positive selection in this fish reveals potential candidates to explain its compressed lifespan. Several aging genes are under positive selection in this short-lived fish and long-lived species, raising the intriguing possibility that the same gene could underlie evolution of both compressed and extended lifespans. Comparative genomics and linkage analysis identify candidate genes associated with lifespan differences between various turquoise killifish strains. Remarkably, these genes are clustered on the sex chromosome, suggesting that short lifespan might have co-evolved with sex determination. Our study provides insights into the evolutionary forces that shape lifespan in nature.


Asunto(s)
Evolución Biológica , Peces Killi/genética , Envejecimiento , Animales , ADN Helicasas/genética , Genoma , Humanos , Longevidad , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Selección Genética
7.
Cell ; 159(4): 709-13, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25417146

RESUMEN

Mammalian aging can be delayed with genetic, dietary, and pharmacologic approaches. Given that the elderly population is dramatically increasing and that aging is the greatest risk factor for a majority of chronic diseases driving both morbidity and mortality, it is critical to expand geroscience research directed at extending human healthspan.


Asunto(s)
Envejecimiento/fisiología , Enfermedad Crónica , Envejecimiento/patología , Animales , Investigación Biomédica , Epigénesis Genética , Interacción Gen-Ambiente , Humanos
8.
Cell ; 158(3): 673-88, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083876

RESUMEN

Trimethylation of histone H3 at lysine 4 (H3K4me3) is a chromatin modification known to mark the transcription start sites of active genes. Here, we show that H3K4me3 domains that spread more broadly over genes in a given cell type preferentially mark genes that are essential for the identity and function of that cell type. Using the broadest H3K4me3 domains as a discovery tool in neural progenitor cells, we identify novel regulators of these cells. Machine learning models reveal that the broadest H3K4me3 domains represent a distinct entity, characterized by increased marks of elongation. The broadest H3K4me3 domains also have more paused polymerase at their promoters, suggesting a unique transcriptional output. Indeed, genes marked by the broadest H3K4me3 domains exhibit enhanced transcriptional consistency and [corrected] increased transcriptional levels, and perturbation of H3K4me3 breadth leads to changes in transcriptional consistency. Thus, H3K4me3 breadth contains information that could ensure transcriptional precision at key cell identity/function genes.


Asunto(s)
Células/metabolismo , Código de Histonas , Histonas/metabolismo , Transcripción Genética , Animales , Inteligencia Artificial , Genómica , Humanos , Lisina/metabolismo , Metilación , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , ARN Polimerasa II/metabolismo
9.
Cell ; 155(3): 621-35, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24243019

RESUMEN

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Asunto(s)
Reprogramación Celular , Embrión de Mamíferos/citología , Fibroblastos/citología , Redes Reguladoras de Genes , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
10.
Mol Cell ; 78(2): 329-345.e9, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32268122

RESUMEN

Neural stem and progenitor cells (NSPCs) are critical for continued cellular replacement in the adult brain. Lifelong maintenance of a functional NSPC pool necessitates stringent mechanisms to preserve a pristine proteome. We find that the NSPC chaperone network robustly maintains misfolded protein solubility and stress resilience through high levels of the ATP-dependent chaperonin TRiC/CCT. Strikingly, NSPC differentiation rewires the cellular chaperone network, reducing TRiC/CCT levels and inducing those of the ATP-independent small heat shock proteins (sHSPs). This switches the proteostasis strategy in neural progeny cells to promote sequestration of misfolded proteins into protective inclusions. The chaperone network of NSPCs is more effective than that of differentiated cells, leading to improved management of proteotoxic stress and amyloidogenic proteins. However, NSPC proteostasis is impaired by brain aging. The less efficient chaperone network of differentiated neural progeny may contribute to their enhanced susceptibility to neurodegenerative diseases characterized by aberrant protein misfolding and aggregation.


Asunto(s)
Envejecimiento/genética , Chaperonas Moleculares/genética , Células-Madre Neurales/metabolismo , Agregación Patológica de Proteínas/genética , Adenosina Trifosfato/genética , Envejecimiento/patología , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Chaperoninas/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Ratones , Chaperonas Moleculares/metabolismo , Células-Madre Neurales/patología , Pliegue de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Células Madre/metabolismo , Células Madre/patología
11.
Nat Methods ; 21(3): 444-454, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38347138

RESUMEN

Whole-transcriptome spatial profiling of genes at single-cell resolution remains a challenge. To address this limitation, spatial gene expression prediction methods have been developed to infer the spatial expression of unmeasured transcripts, but the quality of these predictions can vary greatly. Here we present Transcript Imputation with Spatial Single-cell Uncertainty Estimation (TISSUE) as a general framework for estimating uncertainty for spatial gene expression predictions and providing uncertainty-aware methods for downstream inference. Leveraging conformal inference, TISSUE provides well-calibrated prediction intervals for predicted expression values across 11 benchmark datasets. Moreover, it consistently reduces the false discovery rate for differential gene expression analysis, improves clustering and visualization of predicted spatial transcriptomics and improves the performance of supervised learning models trained on predicted gene expression profiles. Applying TISSUE to a MERFISH spatial transcriptomics dataset of the adult mouse subventricular zone, we identified subtypes within the neural stem cell lineage and developed subtype-specific regional classifiers.


Asunto(s)
Perfilación de la Expresión Génica , Células-Madre Neurales , Animales , Ratones , Incertidumbre , Benchmarking , Análisis por Conglomerados , Transcriptoma , Análisis de la Célula Individual
13.
Nat Rev Mol Cell Biol ; 16(10): 593-610, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26373265

RESUMEN

Ageing is affected by both genetic and non-genetic factors. Here, we review the chromatin-based epigenetic changes that occur during ageing, the role of chromatin modifiers in modulating lifespan and the importance of epigenetic signatures as biomarkers of ageing. We also discuss how epigenome remodelling by environmental stimuli affects several aspects of transcription and genomic stability, with important consequences for longevity, and outline epigenetic differences between the 'mortal soma' and the 'immortal germ line'. Finally, we discuss the inheritance of characteristics of ageing and potential chromatin-based strategies to delay or reverse hallmarks of ageing or age-related diseases.


Asunto(s)
Envejecimiento/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Interacción Gen-Ambiente , Inestabilidad Genómica , Envejecimiento/genética , Envejecimiento/patología , Animales , Biomarcadores/metabolismo , Cromatina/genética , Cromatina/patología , Humanos
15.
Nature ; 574(7779): 553-558, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31645721

RESUMEN

Age-associated chronic inflammation (inflammageing) is a central hallmark of ageing1, but its influence on specific cells remains largely unknown. Fibroblasts are present in most tissues and contribute to wound healing2,3. They are also the most widely used cell type for reprogramming to induced pluripotent stem (iPS) cells, a process that has implications for regenerative medicine and rejuvenation strategies4. Here we show that fibroblast cultures from old mice secrete inflammatory cytokines and exhibit increased variability in the efficiency of iPS cell reprogramming between mice. Variability between individuals is emerging as a feature of old age5-8, but the underlying mechanisms remain unknown. To identify drivers of this variability, we performed multi-omics profiling of fibroblast cultures from young and old mice that have different reprogramming efficiencies. This approach revealed that fibroblast cultures from old mice contain 'activated fibroblasts' that secrete inflammatory cytokines, and that the proportion of activated fibroblasts in a culture correlates with the reprogramming efficiency of that culture. Experiments in which conditioned medium was swapped between cultures showed that extrinsic factors secreted by activated fibroblasts underlie part of the variability between mice in reprogramming efficiency, and we have identified inflammatory cytokines, including TNF, as key contributors. Notably, old mice also exhibited variability in wound healing rate in vivo. Single-cell RNA-sequencing analysis identified distinct subpopulations of fibroblasts with different cytokine expression and signalling in the wounds of old mice with slow versus fast healing rates. Hence, a shift in fibroblast composition, and the ratio of inflammatory cytokines that they secrete, may drive the variability between mice in reprogramming in vitro and influence wound healing rate in vivo. This variability may reflect distinct stochastic ageing trajectories between individuals, and could help in developing personalized strategies to improve iPS cell generation and wound healing in elderly individuals.


Asunto(s)
Envejecimiento/metabolismo , Reprogramación Celular , Senescencia Celular/fisiología , Fibroblastos/metabolismo , Cicatrización de Heridas , Animales , Línea Celular , Reprogramación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Mediadores de Inflamación/metabolismo , Judíos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Transducción de Señal/efectos de los fármacos , Análisis de la Célula Individual , Procesos Estocásticos , Factores de Tiempo , Cicatrización de Heridas/efectos de los fármacos
16.
Nature ; 571(7764): 205-210, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31270459

RESUMEN

The mammalian brain contains neurogenic niches that comprise neural stem cells and other cell types. Neurogenic niches become less functional with age, but how they change during ageing remains unclear. Here we perform single-cell RNA sequencing of young and old neurogenic niches in mice. The analysis of 14,685 single-cell transcriptomes reveals a decrease in activated neural stem cells, changes in endothelial cells and microglia, and an infiltration of T cells in old neurogenic niches. T cells in old brains are clonally expanded and are generally distinct from those in old blood, which suggests that they may experience specific antigens. T cells in old brains also express interferon-γ, and the subset of neural stem cells that has a high interferon response shows decreased proliferation in vivo. We find that T cells can inhibit the proliferation of neural stem cells in co-cultures and in vivo, in part by secreting interferon-γ. Our study reveals an interaction between T cells and neural stem cells in old brains, opening potential avenues through which to counteract age-related decline in brain function.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/citología , Movimiento Celular , Células-Madre Neurales/citología , Neurogénesis , Análisis de la Célula Individual , Nicho de Células Madre/fisiología , Linfocitos T/citología , Animales , Sangre , Proliferación Celular , Células Clonales/citología , Técnicas de Cocultivo , Células Endoteliales/citología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Análisis de Secuencia de ARN , Transducción de Señal , Linfocitos T/metabolismo , Transcriptoma/genética
17.
Mol Cell ; 62(5): 728-44, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259204

RESUMEN

During aging, the mechanisms that normally maintain health and stress resistance strikingly decline, resulting in decrepitude, frailty, and ultimately death. Exactly when and how this decline occurs is unknown. Changes in transcriptional networks and chromatin state lie at the heart of age-dependent decline. These epigenomic changes are not only observed during aging but also profoundly affect cellular function and stress resistance, thereby contributing to the progression of aging. We propose that the dysregulation of transcriptional and chromatin networks is a crucial component of aging. Understanding age-dependent epigenomic changes will yield key insights into how aging begins and progresses and should lead to the development of new therapeutics that delay or even reverse aging and age-related diseases.


Asunto(s)
Envejecimiento/genética , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Factores de Transcripción/genética , Transcripción Genética , Factores de Edad , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Metilación de ADN , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Epigenómica/métodos , Inestabilidad Genómica , Genotipo , Histonas/metabolismo , Humanos , Mitocondrias/metabolismo , Estado Nutricional , Fenotipo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Telómero/genética , Telómero/metabolismo , Factores de Transcripción/metabolismo
18.
Nature ; 544(7649): 185-190, 2017 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-28379943

RESUMEN

Chromatin and metabolic states both influence lifespan, but how they interact in lifespan regulation is largely unknown. The COMPASS chromatin complex, which trimethylates lysine 4 on histone H3 (H3K4me3), regulates lifespan in Caenorhabditis elegans. However, the mechanism by which H3K4me3 modifiers affect longevity, and whether this mechanism involves metabolic changes, remain unclear. Here we show that a deficiency in H3K4me3 methyltransferase, which extends lifespan, promotes fat accumulation in worms with a specific enrichment of mono-unsaturated fatty acids (MUFAs). This fat metabolism switch in H3K4me3 methyltransferase-deficient worms is mediated at least in part by the downregulation of germline targets, including S6 kinase, and by the activation of an intestinal transcriptional network that upregulates delta-9 fatty acid desaturases. Notably, the accumulation of MUFAs is necessary for the lifespan extension of H3K4me3 methyltransferase-deficient worms, and dietary MUFAs are sufficient to extend lifespan. Given the conservation of lipid metabolism, dietary or endogenous MUFAs could extend lifespan and healthspan in other species, including mammals.


Asunto(s)
Caenorhabditis elegans/fisiología , Grasas de la Dieta/farmacología , Ácidos Grasos Insaturados/metabolismo , Histonas/metabolismo , Longevidad/efectos de los fármacos , Longevidad/fisiología , Lisina/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Animales , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/metabolismo , Regulación hacia Abajo , Ácido Graso Desaturasas/genética , Ácido Graso Desaturasas/metabolismo , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/farmacología , Regulación Enzimológica de la Expresión Génica , Células Germinativas/enzimología , Células Germinativas/metabolismo , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Mucosa Intestinal/metabolismo , Intestinos/enzimología , Metabolismo de los Lípidos/efectos de los fármacos , Metilación , Proteínas Quinasas S6 Ribosómicas 70-kDa/deficiencia , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Estearoil-CoA Desaturasa , Regulación hacia Arriba
19.
Nature ; 550(7675): 249-254, 2017 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-29022589

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing is a conserved post-transcriptional mechanism mediated by ADAR enzymes that diversifies the transcriptome by altering selected nucleotides in RNA molecules. Although many editing sites have recently been discovered, the extent to which most sites are edited and how the editing is regulated in different biological contexts are not fully understood. Here we report dynamic spatiotemporal patterns and new regulators of RNA editing, discovered through an extensive profiling of A-to-I RNA editing in 8,551 human samples (representing 53 body sites from 552 individuals) from the Genotype-Tissue Expression (GTEx) project and in hundreds of other primate and mouse samples. We show that editing levels in non-repetitive coding regions vary more between tissues than editing levels in repetitive regions. Globally, ADAR1 is the primary editor of repetitive sites and ADAR2 is the primary editor of non-repetitive coding sites, whereas the catalytically inactive ADAR3 predominantly acts as an inhibitor of editing. Cross-species analysis of RNA editing in several tissues revealed that species, rather than tissue type, is the primary determinant of editing levels, suggesting stronger cis-directed regulation of RNA editing for most sites, although the small set of conserved coding sites is under stronger trans-regulation. In addition, we curated an extensive set of ADAR1 and ADAR2 targets and showed that many editing sites display distinct tissue-specific regulation by the ADAR enzymes in vivo. Further analysis of the GTEx data revealed several potential regulators of editing, such as AIMP2, which reduces editing in muscles by enhancing the degradation of the ADAR proteins. Collectively, our work provides insights into the complex cis- and trans-regulation of A-to-I editing.


Asunto(s)
Adenosina Desaminasa , Primates/genética , Edición de ARN/genética , Proteínas de Unión al ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Femenino , Genotipo , Células HEK293 , Humanos , Masculino , Ratones , Músculos/metabolismo , Proteínas Nucleares/metabolismo , Especificidad de Órganos/genética , Proteolisis , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis Espacio-Temporal , Especificidad de la Especie , Transcriptoma/genética
20.
Mol Cell ; 59(4): 515-6, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26295957

RESUMEN

In this issue of Molecular Cell, Labbadia and Morimoto (2015) show that there is a precipitous decline in stress resistance at the onset of reproduction in C. elegans and that this transition is regulated by changes in repressive chromatin marks.


Asunto(s)
Caenorhabditis elegans/fisiología , Respuesta al Choque Térmico , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA