Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(6): 1453-67, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26046444

RESUMEN

Resetting of the epigenome in human primordial germ cells (hPGCs) is critical for development. We show that the transcriptional program of hPGCs is distinct from that in mice, with co-expression of somatic specifiers and naive pluripotency genes TFCP2L1 and KLF4. This unique gene regulatory network, established by SOX17 and BLIMP1, drives comprehensive germline DNA demethylation by repressing DNA methylation pathways and activating TET-mediated hydroxymethylation. Base-resolution methylome analysis reveals progressive DNA demethylation to basal levels in week 5-7 in vivo hPGCs. Concurrently, hPGCs undergo chromatin reorganization, X reactivation, and imprint erasure. Despite global hypomethylation, evolutionarily young and potentially hazardous retroelements, like SVA, remain methylated. Remarkably, some loci associated with metabolic and neurological disorders are also resistant to DNA demethylation, revealing potential for transgenerational epigenetic inheritance that may have phenotypic consequences. We provide comprehensive insight on early human germline transcriptional network and epigenetic reprogramming that subsequently impacts human development and disease.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Genoma Humano , Células Germinativas/metabolismo , Animales , Metilación de ADN , Embrión de Mamíferos/metabolismo , Femenino , Humanos , Factor 4 Similar a Kruppel , Masculino , Ratones , Regiones Promotoras Genéticas , Retroelementos
2.
Cell ; 160(1-2): 253-68, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25543152

RESUMEN

Specification of primordial germ cells (PGCs) marks the beginning of the totipotent state. However, without a tractable experimental model, the mechanism of human PGC (hPGC) specification remains unclear. Here, we demonstrate specification of hPGC-like cells (hPGCLCs) from germline competent pluripotent stem cells. The characteristics of hPGCLCs are consistent with the embryonic hPGCs and a germline seminoma that share a CD38 cell-surface marker, which collectively defines likely progression of the early human germline. Remarkably, SOX17 is the key regulator of hPGC-like fate, whereas BLIMP1 represses endodermal and other somatic genes during specification of hPGCLCs. Notable mechanistic differences between mouse and human PGC specification could be attributed to their divergent embryonic development and pluripotent states, which might affect other early cell-fate decisions. We have established a foundation for future studies on resetting of the epigenome in hPGCLCs and hPGCs for totipotency and the transmission of genetic and epigenetic information.


Asunto(s)
Diferenciación Celular , Células Germinativas/citología , Factores de Transcripción SOXF/metabolismo , ADP-Ribosil Ciclasa 1/metabolismo , Animales , Línea Celular Tumoral , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Células Germinativas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Represoras/metabolismo , Seminoma/metabolismo , Análisis de Secuencia de ARN
3.
Cell ; 153(2): 335-47, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582324

RESUMEN

Factors that sustain self-renewal of mouse embryonic stem cells (ESCs) are well described. In contrast, the machinery regulating exit from pluripotency is ill defined. In a large-scale small interfering RNA (siRNA) screen, we found that knockdown of the tumor suppressors Folliculin (Flcn) and Tsc2 prevent ESC commitment. Tsc2 lies upstream of mammalian target of rapamycin (mTOR), whereas Flcn acts downstream and in parallel. Flcn with its interaction partners Fnip1 and Fnip2 drives differentiation by restricting nuclear localization and activity of the bHLH transcription factor Tfe3. Conversely, enforced nuclear Tfe3 enables ESCs to withstand differentiation conditions. Genome-wide location and functional analyses showed that Tfe3 directly integrates into the pluripotency circuitry through transcriptional regulation of Esrrb. These findings identify a cell-intrinsic rheostat for destabilizing ground-state pluripotency to allow lineage commitment. Congruently, stage-specific subcellular relocalization of Tfe3 suggests that Flcn-Fnip1/2 contributes to developmental progression of the pluripotent epiblast in vivo.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Redes Reguladoras de Genes , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Células Madre Embrionarias/metabolismo , Estrona/genética , Estrona/metabolismo , Ratones , Ratones Endogámicos C57BL , Serina-Treonina Quinasas TOR/metabolismo
4.
Nature ; 605(7911): 722-727, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35545673

RESUMEN

Cellular diversification is critical for specialized functions of the brain including learning and memory1. Single-cell RNA sequencing facilitates transcriptomic profiling of distinct major types of neuron2-4, but the divergence of transcriptomic profiles within a neuronal population and their link to function remain poorly understood. Here we isolate nuclei tagged5 in specific cell types followed by single-nucleus RNA sequencing to profile Purkinje neurons and map their responses to motor activity and learning. We find that two major subpopulations of Purkinje neurons, identified by expression of the genes Aldoc and Plcb4, bear distinct transcriptomic features. Plcb4+, but not Aldoc+, Purkinje neurons exhibit robust plasticity of gene expression in mice subjected to sensorimotor and learning experience. In vivo calcium imaging and optogenetic perturbation reveal that Plcb4+ Purkinje neurons have a crucial role in associative learning. Integrating single-nucleus RNA sequencing datasets with weighted gene co-expression network analysis uncovers a learning gene module that includes components of FGFR2 signalling in Plcb4+ Purkinje neurons. Knockout of Fgfr2 in Plcb4+ Purkinje neurons in mice using CRISPR disrupts motor learning. Our findings define how diversification of Purkinje neurons is linked to their responses in motor learning and provide a foundation for understanding their differential vulnerability to neurological disorders.


Asunto(s)
Células de Purkinje , Transcriptoma , Animales , Cerebelo , Aprendizaje/fisiología , Ratones , Ratones Noqueados , Plasticidad Neuronal/genética , Neuronas/fisiología , Células de Purkinje/metabolismo , Transcriptoma/genética
5.
Nature ; 607(7919): 593-603, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35768510

RESUMEN

Aggressive and metastatic cancers show enhanced metabolic plasticity1, but the precise underlying mechanisms of this remain unclear. Here we show how two NOP2/Sun RNA methyltransferase 3 (NSUN3)-dependent RNA modifications-5-methylcytosine (m5C) and its derivative 5-formylcytosine (f5C) (refs.2-4)-drive the translation of mitochondrial mRNA to power metastasis. Translation of mitochondrially encoded subunits of the oxidative phosphorylation complex depends on the formation of m5C at position 34 in mitochondrial tRNAMet. m5C-deficient human oral cancer cells exhibit increased levels of glycolysis and changes in their mitochondrial function that do not affect cell viability or primary tumour growth in vivo; however, metabolic plasticity is severely impaired as mitochondrial m5C-deficient tumours do not metastasize efficiently. We discovered that CD36-dependent non-dividing, metastasis-initiating tumour cells require mitochondrial m5C to activate invasion and dissemination. Moreover, a mitochondria-driven gene signature in patients with head and neck cancer is predictive for metastasis and disease progression. Finally, we confirm that this metabolic switch that allows the metastasis of tumour cells can be pharmacologically targeted through the inhibition of mitochondrial mRNA translation in vivo. Together, our results reveal that site-specific mitochondrial RNA modifications could be therapeutic targets to combat metastasis.


Asunto(s)
5-Metilcitosina , Citosina/análogos & derivados , Glucólisis , Mitocondrias , Metástasis de la Neoplasia , Fosforilación Oxidativa , ARN Mitocondrial , 5-Metilcitosina/biosíntesis , 5-Metilcitosina/metabolismo , Antígenos CD36 , Supervivencia Celular , Citosina/metabolismo , Progresión de la Enfermedad , Glucólisis/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Fosforilación Oxidativa/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo , ARN de Transferencia de Metionina/genética , ARN de Transferencia de Metionina/metabolismo
6.
Development ; 151(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38602479

RESUMEN

Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFBs) and a stable but poorly described population of lipid-rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFBs). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single-cell RNA sequencing and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a MyoFB differentiation program that is distinct from other mesenchymal cell types and increases the known repertoire of mesenchymal cell types in the neonatal lung.


Asunto(s)
Animales Recién Nacidos , Diferenciación Celular , Pulmón , Miofibroblastos , Animales , Miofibroblastos/metabolismo , Miofibroblastos/citología , Ratones , Pulmón/citología , Pulmón/embriología , Pulmón/metabolismo , Linaje de la Célula , Organogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo
7.
Mol Cell ; 71(1): 56-72.e4, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-30008319

RESUMEN

Chromatin remodeling complexes play essential roles in metazoan development through widespread control of gene expression, but the precise molecular mechanisms by which they do this in vivo remain ill defined. Using an inducible system with fine temporal resolution, we show that the nucleosome remodeling and deacetylation (NuRD) complex controls chromatin architecture and the protein binding repertoire at regulatory regions during cell state transitions. This is primarily exerted through its nucleosome remodeling activity while deacetylation at H3K27 follows changes in gene expression. Additionally, NuRD activity influences association of RNA polymerase II at transcription start sites and subsequent nascent transcript production, thereby guiding the establishment of lineage-appropriate transcriptional programs. These findings provide a detailed molecular picture of genome-wide modulation of lineage-specific transcription by an essential chromatin remodeling complex as well as insight into the orchestration of molecular events involved in transcriptional transitions in vivo. VIDEO ABSTRACT.


Asunto(s)
Regulación de la Expresión Génica , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Nucleosomas/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Acetilación , Animales , Línea Celular , Histonas/genética , Histonas/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Nucleosomas/genética , ARN Polimerasa II/genética , Sitio de Iniciación de la Transcripción
8.
RNA Biol ; 21(1): 1-18, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38566310

RESUMEN

RNA modifications, including N-7-methylguanosine (m7G), are pivotal in governing RNA stability and gene expression regulation. The accurate detection of internal m7G modifications is of paramount significance, given recent associations between altered m7G deposition and elevated expression of the methyltransferase METTL1 in various human cancers. The development of robust m7G detection techniques has posed a significant challenge in the field of epitranscriptomics. In this study, we introduce two methodologies for the global and accurate identification of m7G modifications in human RNA. We introduce borohydride reduction sequencing (Bo-Seq), which provides base resolution mapping of m7G modifications. Bo-Seq achieves exceptional performance through the optimization of RNA depurination and scission, involving the strategic use of high concentrations of NaBH4, neutral pH and the addition of 7-methylguanosine monophosphate (m7GMP) during the reducing reaction. Notably, compared to NaBH4-based methods, Bo-Seq enhances the m7G detection performance, and simplifies the detection process, eliminating the necessity for intricate chemical steps and reducing the protocol duration. In addition, we present an antibody-based approach, which enables the assessment of m7G relative levels across RNA molecules and biological samples, however it should be used with caution due to limitations associated with variations in antibody quality between batches. In summary, our novel approaches address the pressing need for reliable and accessible methods to detect RNA m7G methylation in human cells. These advancements hold the potential to catalyse future investigations in the critical field of epitranscriptomics, shedding light on the complex regulatory roles of m7G in gene expression and its implications in cancer biology.


Asunto(s)
Guanosina/análogos & derivados , Nucleótidos , ARN , Humanos , ARN/química , Nucleótidos/metabolismo , Metilación , Metiltransferasas/genética , Procesamiento Postranscripcional del ARN
9.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34518230

RESUMEN

Genome remethylation is essential for mammalian development but specific reasons are unclear. Here we examined embryonic stem (ES) cell fate in the absence of de novo DNA methyltransferases. We observed that ES cells deficient for both Dnmt3a and Dnmt3b are rapidly eliminated from chimeras. On further investigation we found that in vivo and in vitro the formative pluripotency transition is derailed toward production of trophoblast. This aberrant trajectory is associated with failure to suppress activation of Ascl2Ascl2 encodes a bHLH transcription factor expressed in the placenta. Misexpression of Ascl2 in ES cells provokes transdifferentiation to trophoblast-like cells. Conversely, Ascl2 deletion rescues formative transition of Dnmt3a/b mutants and improves contribution to chimeric epiblast. Thus, de novo DNA methylation safeguards against ectopic activation of Ascl2 However, Dnmt3a/b-deficient cells remain defective in ongoing embryogenesis. We surmise that multiple developmental transitions may be secured by DNA methylation silencing potentially disruptive genes.


Asunto(s)
Metilación de ADN/genética , Células Madre Embrionarias/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , Desarrollo Embrionario/genética , Ratones , Trofoblastos/fisiología , ADN Metiltransferasa 3B
10.
Mol Cancer ; 22(1): 119, 2023 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-37516825

RESUMEN

Newly growing evidence highlights the essential role that epitranscriptomic marks play in the development of many cancers; however, little is known about the role and implications of altered epitranscriptome deposition in prostate cancer. Here, we show that the transfer RNA N7-methylguanosine (m7G) transferase METTL1 is highly expressed in primary and advanced prostate tumours. Mechanistically, we find that METTL1 depletion causes the loss of m7G tRNA methylation and promotes the biogenesis of a novel class of small non-coding RNAs derived from 5'tRNA fragments. 5'tRNA-derived small RNAs steer translation control to favour the synthesis of key regulators of tumour growth suppression, interferon pathway, and immune effectors. Knockdown of Mettl1 in prostate cancer preclinical models increases intratumoural infiltration of pro-inflammatory immune cells and enhances responses to immunotherapy. Collectively, our findings reveal a therapeutically actionable role of METTL1-directed m7G tRNA methylation in cancer cell translation control and tumour biology.


Asunto(s)
Carcinogénesis , Neoplasias de la Próstata , Masculino , Humanos , Carcinogénesis/genética , Transformación Celular Neoplásica , Neoplasias de la Próstata/genética , Transcripción Genética , Procesamiento Postranscripcional del ARN , Metiltransferasas/genética
11.
EMBO J ; 38(12)2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31036553

RESUMEN

Multiprotein chromatin remodelling complexes show remarkable conservation of function amongst metazoans, even though components present in invertebrates are often found as multiple paralogous proteins in vertebrate complexes. In some cases, these paralogues specify distinct biochemical and/or functional activities in vertebrate cells. Here, we set out to define the biochemical and functional diversity encoded by one such group of proteins within the mammalian Nucleosome Remodelling and Deacetylation (NuRD) complex: Mta1, Mta2 and Mta3. We find that, in contrast to what has been described in somatic cells, MTA proteins are not mutually exclusive within embryonic stem (ES) cell NuRD and, despite subtle differences in chromatin binding and biochemical interactions, serve largely redundant functions. ES cells lacking all three MTA proteins exhibit complete NuRD loss of function and are viable, allowing us to identify a previously unreported function for NuRD in reducing transcriptional noise, which is essential for maintaining a proper differentiation trajectory during early stages of lineage commitment.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/fisiología , Transcripción Genética , Animales , Células Cultivadas , Reprogramación Celular/genética , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/fisiología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Proteínas Represoras/genética , Proteínas Represoras/fisiología , Relación Señal-Ruido , Transactivadores/genética , Transactivadores/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transcripción Genética/fisiología
12.
Nature ; 546(7658): 416-420, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28607482

RESUMEN

Human primordial germ cells (hPGCs), the precursors of sperm and eggs, originate during weeks 2-3 of early post-implantation development. Using in vitro models of hPGC induction, recent studies have suggested that there are marked mechanistic differences in the specification of human and mouse PGCs. This may be due in part to the divergence in their pluripotency networks and early post-implantation development. As early human embryos are not accessible for direct study, we considered alternatives including porcine embryos that, as in humans, develop as bilaminar embryonic discs. Here we show that porcine PGCs originate from the posterior pre-primitive-streak competent epiblast by sequential upregulation of SOX17 and BLIMP1 in response to WNT and BMP signalling. We use this model together with human and monkey in vitro models simulating peri-gastrulation development to show the conserved principles of epiblast development for competency for primordial germ cell fate. This process is followed by initiation of the epigenetic program and regulated by a balanced SOX17-BLIMP1 gene dosage. Our combinatorial approach using human, porcine and monkey in vivo and in vitro models provides synthetic insights into early human development.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Células Germinativas/citología , Macaca fascicularis/embriología , Modelos Biológicos , Células Madre Pluripotentes/citología , Porcinos/embriología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Linaje de la Célula , Cuerpos Embrioides/citología , Epigénesis Genética , Femenino , Gastrulación , Dosificación de Gen , Células Germinativas/metabolismo , Estratos Germinativos/citología , Humanos , Técnicas In Vitro , Masculino , Modelos Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Línea Primitiva/citología , Proteínas Represoras/genética , Factores de Transcripción SOXF/genética , Vía de Señalización Wnt
13.
Nucleic Acids Res ; 49(2): 1006-1022, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33330931

RESUMEN

The highly abundant N6-methyladenosine (m6A) RNA modification affects most aspects of mRNA function, yet the precise function of the rarer 5-methylcytidine (m5C) remains largely unknown. Here, we map m5C in the human transcriptome using methylation-dependent individual-nucleotide resolution cross-linking and immunoprecipitation (miCLIP) combined with RNA bisulfite sequencing. We identify NSUN6 as a methyltransferase with strong substrate specificity towards mRNA. NSUN6 primarily targeted three prime untranslated regions (3'UTR) at the consensus sequence motif CTCCA, located in loops of hairpin structures. Knockout and rescue experiments revealed enhanced mRNA and translation levels when NSUN6-targeted mRNAs were methylated. Ribosome profiling further demonstrated that NSUN6-specific methylation correlated with translation termination. While NSUN6 was dispensable for mouse embryonic development, it was down-regulated in human tumours and high expression of NSUN6 indicated better patient outcome of certain cancer types. In summary, our study identifies NSUN6 as a methyltransferase targeting mRNA, potentially as part of a quality control mechanism involved in translation termination fidelity.


Asunto(s)
Citidina/análogos & derivados , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARNt Metiltransferasas/metabolismo , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Línea Celular Tumoral , Uso de Codones , Secuencia de Consenso , Citidina/metabolismo , Células Madre Embrionarias , Técnicas de Inactivación de Genes , Genes Reporteros , Células HEK293 , Humanos , Inmunoprecipitación , Metilación , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , ARN Mensajero/genética , Transcriptoma , ARNt Metiltransferasas/deficiencia
14.
PLoS Biol ; 17(6): e3000297, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31199786

RESUMEN

Posttranscriptional modifications in transfer RNA (tRNA) are often critical for normal development because they adapt protein synthesis rates to a dynamically changing microenvironment. However, the precise cellular mechanisms linking the extrinsic stimulus to the intrinsic RNA modification pathways remain largely unclear. Here, we identified the cytosine-5 RNA methyltransferase NSUN2 as a sensor for external stress stimuli. Exposure to oxidative stress efficiently repressed NSUN2, causing a reduction of methylation at specific tRNA sites. Using metabolic profiling, we showed that loss of tRNA methylation captured cells in a distinct catabolic state. Mechanistically, loss of NSUN2 altered the biogenesis of tRNA-derived noncoding fragments (tRFs) in response to stress, leading to impaired regulation of protein synthesis. The intracellular accumulation of a specific subset of tRFs correlated with the dynamic repression of global protein synthesis. Finally, NSUN2-driven RNA methylation was functionally required to adapt cell cycle progression to the early stress response. In summary, we revealed that changes in tRNA methylation profiles were sufficient to specify cellular metabolic states and efficiently adapt protein synthesis rates to cell stress.


Asunto(s)
ADN-Citosina Metilasas/metabolismo , Metiltransferasas/metabolismo , Animales , Línea Celular , Citosina/metabolismo , Metilación de ADN/fisiología , ADN-Citosina Metilasas/fisiología , Humanos , Ratones , Estrés Oxidativo/fisiología , Biosíntesis de Proteínas/fisiología , ARN/metabolismo , ARN de Transferencia/metabolismo
15.
Nat Rev Genet ; 17(10): 585-600, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27573372

RESUMEN

Primordial germ cells (PGCs), the precursors of sperm and eggs, are established in perigastrulation-stage embryos in mammals. Signals from extra-embryonic tissues induce a unique gene regulatory network in germline-competent cells for PGC specification. This network also initiates comprehensive epigenome resetting, including global DNA demethylation and chromatin reorganization. Mouse germline development has been studied extensively, but the extent to which such knowledge applies to humans was unclear. Here, we review the latest advances in human PGC specification and epigenetic reprogramming. The overall developmental dynamics of human and mouse germline cells appear to be similar, but there are crucial mechanistic differences in PGC specification, reflecting divergence in the regulation of pluripotency and early development.


Asunto(s)
Metilación de ADN , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Germinativas , Animales , Humanos , Ratones , Transducción de Señal
16.
Nature ; 534(7607): 335-40, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27306184

RESUMEN

Whether protein synthesis and cellular stress response pathways interact to control stem cell function is currently unknown. Here we show that mouse skin stem cells synthesize less protein than their immediate progenitors in vivo, even when forced to proliferate. Our analyses reveal that activation of stress response pathways drives both a global reduction of protein synthesis and altered translational programmes that together promote stem cell functions and tumorigenesis. Mechanistically, we show that inhibition of post-transcriptional cytosine-5 methylation locks tumour-initiating cells in this distinct translational inhibition programme. Paradoxically, this inhibition renders stem cells hypersensitive to cytotoxic stress, as tumour regeneration after treatment with 5-fluorouracil is blocked. Thus, stem cells must revoke translation inhibition pathways to regenerate a tissue or tumour.


Asunto(s)
Biosíntesis de Proteínas , Células Madre/fisiología , Estrés Fisiológico , Animales , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citosina/metabolismo , Femenino , Fluorouracilo/farmacología , Folículo Piloso/citología , Folículo Piloso/metabolismo , Humanos , Masculino , Metilación , Metiltransferasas/deficiencia , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Regeneración , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Células Madre/citología , Estrés Fisiológico/genética
17.
Nature ; 529(7586): 403-407, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26751055

RESUMEN

Nanog, a core pluripotency factor in the inner cell mass of blastocysts, is also expressed in unipotent primordial germ cells (PGCs) in mice, where its precise role is yet unclear. We investigated this in an in vitro model, in which naive pluripotent embryonic stem (ES) cells cultured in basic fibroblast growth factor (bFGF) and activin A develop as epiblast-like cells (EpiLCs) and gain competence for a PGC-like fate. Consequently, bone morphogenetic protein 4 (BMP4), or ectopic expression of key germline transcription factors Prdm1, Prdm14 and Tfap2c, directly induce PGC-like cells (PGCLCs) in EpiLCs, but not in ES cells. Here we report an unexpected discovery that Nanog alone can induce PGCLCs in EpiLCs, independently of BMP4. We propose that after the dissolution of the naive ES-cell pluripotency network during establishment of EpiLCs, the epigenome is reset for cell fate determination. Indeed, we found genome-wide changes in NANOG-binding patterns between ES cells and EpiLCs, indicating epigenetic resetting of regulatory elements. Accordingly, we show that NANOG can bind and activate enhancers of Prdm1 and Prdm14 in EpiLCs in vitro; BLIMP1 (encoded by Prdm1) then directly induces Tfap2c. Furthermore, while SOX2 and NANOG promote the pluripotent state in ES cells, they show contrasting roles in EpiLCs, as Sox2 specifically represses PGCLC induction by Nanog. This study demonstrates a broadly applicable mechanistic principle for how cells acquire competence for cell fate determination, resulting in the context-dependent roles of key transcription factors during development.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Células Germinativas/citología , Células Germinativas/metabolismo , Estratos Germinativos/citología , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias de Ratones/citología , Factores de Transcripción/genética , Activinas/farmacología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Proteínas de Unión al ADN , Epigénesis Genética , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Estratos Germinativos/efectos de los fármacos , Estratos Germinativos/metabolismo , Proteínas de Homeodominio/antagonistas & inhibidores , Masculino , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Proteína Homeótica Nanog , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Unión Proteica , Proteínas de Unión al ARN , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Factores de Transcripción/metabolismo
18.
Mol Cell ; 56(4): 564-79, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25457166

RESUMEN

Primordial germ cells (PGCs) and preimplantation embryos undergo epigenetic reprogramming, which includes comprehensive DNA demethylation. We found that PRMT5, an arginine methyltransferase, translocates from the cytoplasm to the nucleus during this process. Here we show that conditional loss of PRMT5 in early PGCs causes complete male and female sterility, preceded by the upregulation of LINE1 and IAP transposons as well as activation of a DNA damage response. Similarly, loss of maternal-zygotic PRMT5 also leads to IAP upregulation. PRMT5 is necessary for the repressive H2A/H4R3me2s chromatin modification on LINE1 and IAP transposons in PGCs, directly implicating this modification in transposon silencing during DNA hypomethylation. PRMT5 translocates back to the cytoplasm subsequently, to participate in the previously described PIWI-interacting RNA (piRNA) pathway that promotes transposon silencing via de novo DNA remethylation. Thus, PRMT5 is directly involved in genome defense during preimplantation development and in PGCs at the time of global DNA demethylation.


Asunto(s)
Blastocisto/enzimología , Metilación de ADN , Inestabilidad Genómica , Óvulo/enzimología , Proteína Metiltransferasas/fisiología , Espermatozoides/enzimología , Animales , Apoptosis , Blastocisto/citología , Células Cultivadas , Daño del ADN , Elementos Transponibles de ADN , Desarrollo Embrionario , Células Madre Embrionarias/enzimología , Femenino , Histonas/metabolismo , Masculino , Ratones Transgénicos , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas
19.
Development ; 145(21)2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30413530

RESUMEN

The mouse embryo is the canonical model for mammalian preimplantation development. Recent advances in single cell profiling allow detailed analysis of embryogenesis in other eutherian species, including human, to distinguish conserved from divergent regulatory programs and signalling pathways in the rodent paradigm. Here, we identify and compare transcriptional features of human, marmoset and mouse embryos by single cell RNA-seq. Zygotic genome activation correlates with the presence of polycomb repressive complexes in all three species, while ribosome biogenesis emerges as a predominant attribute in primate embryos, supporting prolonged translation of maternally deposited RNAs. We find that transposable element expression signatures are species, stage and lineage specific. The pluripotency network in the primate epiblast lacks certain regulators that are operative in mouse, but encompasses WNT components and genes associated with trophoblast specification. Sequential activation of GATA6, SOX17 and GATA4 markers of primitive endoderm identity is conserved in primates. Unexpectedly, OTX2 is also associated with primitive endoderm specification in human and non-human primate blastocysts. Our cross-species analysis demarcates both conserved and primate-specific features of preimplantation development, and underscores the molecular adaptability of early mammalian embryogenesis.


Asunto(s)
Callithrix/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Secuencia Conservada/genética , Endodermo/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Estratos Germinativos/metabolismo , Humanos , Ratones , Factores de Transcripción Otx , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/genética , Transcripción Genética
20.
Development ; 144(15): 2748-2763, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28765214

RESUMEN

Much attention has focussed on the conversion of human pluripotent stem cells (PSCs) to a more naïve developmental status. Here we provide a method for resetting via transient histone deacetylase inhibition. The protocol is effective across multiple PSC lines and can proceed without karyotype change. Reset cells can be expanded without feeders with a doubling time of around 24 h. WNT inhibition stabilises the resetting process. The transcriptome of reset cells diverges markedly from that of primed PSCs and shares features with human inner cell mass (ICM). Reset cells activate expression of primate-specific transposable elements. DNA methylation is globally reduced to a level equivalent to that in the ICM and is non-random, with gain of methylation at specific loci. Methylation imprints are mostly lost, however. Reset cells can be re-primed to undergo tri-lineage differentiation and germline specification. In female reset cells, appearance of biallelic X-linked gene transcription indicates reactivation of the silenced X chromosome. On reconversion to primed status, XIST-induced silencing restores monoallelic gene expression. The facile and robust conversion routine with accompanying data resources will enable widespread utilisation, interrogation, and refinement of candidate naïve cells.


Asunto(s)
Elementos Transponibles de ADN/genética , Epigénesis Genética/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Metilación de ADN/genética , Metilación de ADN/fisiología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Citometría de Flujo , Genes Ligados a X/genética , Humanos , Hibridación Fluorescente in Situ , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Inactivación del Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA