Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunity ; 48(6): 1258-1270.e6, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29884461

RESUMEN

Thymus development is critical to the adaptive immune system, yet a comprehensive transcriptional framework capturing thymus organogenesis at single-cell resolution is still needed. We applied single-cell RNA sequencing (RNA-seq) to capture 8 days of thymus development, perturbations of T cell receptor rearrangement, and in vitro organ cultures, producing profiles of 24,279 cells. We resolved transcriptional heterogeneity of developing lymphocytes, and genetic perturbation confirmed T cell identity of conventional and non-conventional lymphocytes. We characterized maturation dynamics of thymic epithelial cells in vivo, classified cell maturation state in a thymic organ culture, and revealed the intrinsic capacity of thymic epithelium to preserve transcriptional regularity despite exposure to exogenous retinoic acid. Finally, by integrating the cell atlas with human genome-wide association study (GWAS) data and autoimmune-disease-related genes, we implicated embryonic thymus-resident cells as possible participants in autoimmune disease etiologies. This resource provides a single-cell transcriptional framework for biological discovery and molecular analysis of thymus organogenesis.


Asunto(s)
Diferenciación Celular/inmunología , Análisis de Secuencia de ARN/métodos , Linfocitos T/inmunología , Timo/embriología , Animales , Enfermedades Autoinmunes/inmunología , Embrión de Mamíferos , Perfilación de la Expresión Génica/métodos , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Organogénesis/inmunología , Linfocitos T/citología , Timo/citología
2.
Nat Methods ; 18(9): 1046-1055, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34480151

RESUMEN

Chromosome conformation capture (3C) assays are used to map chromatin interactions genome-wide. Chromatin interaction maps provide insights into the spatial organization of chromosomes and the mechanisms by which they fold. Hi-C and Micro-C are widely used 3C protocols that differ in key experimental parameters including cross-linking chemistry and chromatin fragmentation strategy. To understand how the choice of experimental protocol determines the ability to detect and quantify aspects of chromosome folding we have performed a systematic evaluation of 3C experimental parameters. We identified optimal protocol variants for either loop or compartment detection, optimizing fragment size and cross-linking chemistry. We used this knowledge to develop a greatly improved Hi-C protocol (Hi-C 3.0) that can detect both loops and compartments relatively effectively. In addition to providing benchmarked protocols, this work produced ultra-deep chromatin interaction maps using Micro-C, conventional Hi-C and Hi-C 3.0 for key cell lines used by the 4D Nucleome project.


Asunto(s)
Cromatina/química , Cromosomas Humanos/química , Reactivos de Enlaces Cruzados/química , Técnicas Genéticas , Línea Celular , Cromatina/metabolismo , Bases de Datos Factuales , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/fisiología , Humanos
3.
Proc Natl Acad Sci U S A ; 116(36): 17970-17979, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31420514

RESUMEN

Single-cell sequencing technologies have revealed an unexpectedly broad repertoire of cells required to mediate complex functions in multicellular organisms. Despite the multiple roles of adipose tissue in maintaining systemic metabolic homeostasis, adipocytes are thought to be largely homogenous with only 2 major subtypes recognized in humans so far. Here we report the existence and characteristics of 4 distinct human adipocyte subtypes, and of their respective mesenchymal progenitors. The phenotypes of these distinct adipocyte subtypes are differentially associated with key adipose tissue functions, including thermogenesis, lipid storage, and adipokine secretion. The transcriptomic signature of "brite/beige" thermogenic adipocytes reveals mechanisms for iron accumulation and protection from oxidative stress, necessary for mitochondrial biogenesis and respiration upon activation. Importantly, this signature is enriched in human supraclavicular adipose tissue, confirming that these cells comprise thermogenic depots in vivo, and explain previous findings of a rate-limiting role of iron in adipose tissue browning. The mesenchymal progenitors that give rise to beige/brite adipocytes express a unique set of cytokines and transcriptional regulators involved in immune cell modulation of adipose tissue browning. Unexpectedly, we also find adipocyte subtypes specialized for high-level expression of the adipokines adiponectin or leptin, associated with distinct transcription factors previously implicated in adipocyte differentiation. The finding of a broad adipocyte repertoire derived from a distinct set of mesenchymal progenitors, and of the transcriptional regulators that can control their development, provides a framework for understanding human adipose tissue function and role in metabolic disease.


Asunto(s)
Adipocitos Beige/metabolismo , Adiponectina/biosíntesis , Leptina/sangre , Células Madre Mesenquimatosas/metabolismo , Termogénesis , Transcriptoma , Adipocitos Beige/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Células Madre Mesenquimatosas/citología
4.
Development ; 141(1): 219-23, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24346702

RESUMEN

The identification of the trans-acting factors and cis-regulatory modules that are involved in human pluripotent stem cell (hPSC) maintenance and differentiation is necessary to dissect the operating regulatory networks in these processes and thereby identify nodes where signal input will direct desired cell fate decisions in vitro or in vivo. To deconvolute these networks, we established a method to influence the differentiation state of hPSCs with a CRISPR-associated catalytically inactive dCas9 fused to an effector domain. In human embryonic stem cells, we find that the dCas9 effectors can exert positive or negative regulation on the expression of developmentally relevant genes, which can influence cell differentiation status when impinging on a key node in the regulatory network that governs the cell state. This system provides a platform for the interrogation of the underlying regulators governing specific differentiation decisions, which can then be employed to direct cellular differentiation down desired pathways.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Asociadas a CRISPR/genética , Sistemas CRISPR-Cas , Diferenciación Celular/genética , Endonucleasas/genética , Redes Reguladoras de Genes/genética , Células Madre Pluripotentes/metabolismo , Activación Transcripcional/genética , Secuencia de Aminoácidos , Proteína 9 Asociada a CRISPR , Linaje de la Célula/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células HEK293 , Humanos , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Factores de Transcripción SOXF/biosíntesis , Transcripción Genética
5.
Dev Cell ; 58(18): 1801-1818.e15, 2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37751684

RESUMEN

Approaches to study human pharyngeal foregut endoderm-a developmental intermediate that is linked to various human syndromes involving pharynx development and organogenesis of tissues such as thymus, parathyroid, and thyroid-have been hampered by scarcity of tissue access and cellular models. We present an efficient stepwise differentiation method to generate human pharyngeal foregut endoderm from pluripotent stem cells. We determine dose and temporal requirements of signaling pathway engagement for optimized differentiation and characterize the differentiation products on cellular and integrated molecular level. We present a computational classification tool, "CellMatch," and transcriptomic classification of differentiation products on an integrated mouse scRNA-seq developmental roadmap confirms cellular maturation. Integrated transcriptomic and chromatin analyses infer differentiation stage-specific gene regulatory networks. Our work provides the method and integrated multiomic resource for the investigation of disease-relevant loci and gene regulatory networks and their role in developmental defects affecting the pharyngeal endoderm and its derivatives.


Asunto(s)
Faringe , Células Madre Pluripotentes , Humanos , Animales , Ratones , Endodermo/metabolismo , Sistema Digestivo , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica
6.
Cell Rep ; 27(3): 708-718.e10, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995470

RESUMEN

Studies in vertebrates have outlined conserved molecular control of definitive endoderm (END) development. However, recent work also shows that key molecular aspects of human END regulation differ even from rodents. Differentiation of human embryonic stem cells (ESCs) to END offers a tractable system to study the molecular basis of normal and defective human-specific END development. Here, we interrogated dynamics in chromatin accessibility during differentiation of ESCs to END, predicting DNA-binding proteins that may drive this cell fate transition. We then combined single-cell RNA-seq with parallel CRISPR perturbations to comprehensively define the loss-of-function phenotype of those factors in END development. Following a few candidates, we revealed distinct impairments in the differentiation trajectories for mediators of TGFß signaling and expose a role for the FOXA2 transcription factor in priming human END competence for human foregut and hepatic END specification. Together, this single-cell functional genomics study provides high-resolution insight on human END development.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , ARN Guía de Kinetoplastida/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Endodermo/citología , Endodermo/metabolismo , Factor Nuclear 3-beta del Hepatocito/antagonistas & inhibidores , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Interferencia de ARN , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo
7.
Nat Biotechnol ; 37(4): 461-468, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30936567

RESUMEN

Recent single-cell RNA-sequencing studies have suggested that cells follow continuous transcriptomic trajectories in an asynchronous fashion during development. However, observations of cell flux along trajectories are confounded with population size effects in snapshot experiments and are therefore hard to interpret. In particular, changes in proliferation and death rates can be mistaken for cell flux. Here we present pseudodynamics, a mathematical framework that reconciles population dynamics with the concepts underlying developmental trajectories inferred from time-series single-cell data. Pseudodynamics models population distribution shifts across trajectories to quantify selection pressure, population expansion, and developmental potentials. Applying this model to time-resolved single-cell RNA-sequencing of T-cell and pancreatic beta cell maturation, we characterize proliferation and apoptosis rates and identify key developmental checkpoints, data inaccessible to existing approaches.


Asunto(s)
Diferenciación Celular/genética , Análisis de Secuencia de ARN/estadística & datos numéricos , Análisis de la Célula Individual/estadística & datos numéricos , Animales , Apoptosis/genética , Biotecnología , Proliferación Celular/genética , Femenino , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Funciones de Verosimilitud , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Factores de Tiempo
8.
Stem Cell Res ; 11(3): 1003-12, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23917481

RESUMEN

Anterior foregut endoderm (AFE) gives rise to therapeutically relevant cell types in tissues such as the esophagus, salivary glands, lung, thymus, parathyroid and thyroid. Despite its importance, reports describing the generation of AFE from pluripotent stem cells (PSCs) by directed differentiation have mainly focused on the Nkx2.1(+) lung and thyroid lineages. Here, we describe a novel protocol to derive a subdomain of AFE, identified by expression of Pax9, from PSCs using small molecules and defined media conditions. We generated a reporter PSC line for isolation and characterization of Pax9(+) AFE cells, which when transplanted in vivo, can form several distinct complex AFE-derived epithelia, including mucosal glands and stratified squamous epithelium. Finally, we show that the directed differentiation protocol can be used to generate AFE from human PSCs. Thus, this work both broadens the range of PSC-derived AFE tissues and creates a platform enabling the study of AFE disorders.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Medios de Cultivo/farmacología , Células Madre Embrionarias/citología , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Factor Nuclear 3-beta del Hepatocito/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX9/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA