Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
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
2.
Nat Commun ; 13(1): 457, 2022 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-35075189

RESUMEN

Maldevelopment of the pharyngeal endoderm, an embryonic tissue critical for patterning of the pharyngeal region and ensuing organogenesis, ultimately contributes to several classes of human developmental syndromes and disorders. Such syndromes are characterized by a spectrum of phenotypes that currently cannot be fully explained by known mutations or genetic variants due to gaps in characterization of critical drivers of normal and dysfunctional development. Despite the disease-relevance of pharyngeal endoderm, we still lack a comprehensive and integrative view of the molecular basis and gene regulatory networks driving pharyngeal endoderm development. To close this gap, we apply transcriptomic and chromatin accessibility single-cell sequencing technologies to generate a multi-omic developmental resource spanning pharyngeal endoderm patterning to the emergence of organ-specific epithelia in the developing mouse embryo. We identify cell-type specific gene regulation, distill GRN models that define developing organ domains, and characterize the role of an immunodeficiency-associated forkhead box transcription factor.


Asunto(s)
Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Faringe/embriología , Transcriptoma , Animales , Cromatina/metabolismo , Endodermo/embriología , Endodermo/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Organogénesis , Faringe/metabolismo , Análisis de la Célula Individual , Timocitos/citología , Timocitos/metabolismo
3.
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
4.
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
5.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...