Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Stem Cell ; 25(3): 388-406.e8, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31422912

RESUMEN

Understanding how cell identity transitions occur and whether there are multiple paths between the same beginning and end states are questions of wide interest. Here we show that acquisition of naive pluripotency can follow transcriptionally and mechanistically distinct routes. Starting from post-implantation epiblast stem cells (EpiSCs), one route advances through a mesodermal state prior to naive pluripotency induction, whereas another transiently resembles the early inner cell mass and correspondingly gains greater developmental potency. These routes utilize distinct signaling networks and transcription factors but subsequently converge on the same naive endpoint, showing surprising flexibility in mechanisms underlying identity transitions and suggesting that naive pluripotency is a multidimensional attractor state. These route differences are reconciled by precise expression of Oct4 as a unifying, essential, and sufficient feature. We propose that fine-tuned regulation of this "transition factor" underpins multidimensional access to naive pluripotency, offering a conceptual framework for understanding cell identity transitions.


Asunto(s)
Masa Celular Interna del Blastocisto/fisiología , Estratos Germinativos/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Línea Celular , Plasticidad de la Célula , Reprogramación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/genética , Transducción de Señal
2.
Exp Hematol ; 76: 1-12.e5, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31326613

RESUMEN

Pluripotent stem cell (PSC) differentiation in vitro represents a powerful and tractable model to study mammalian development and an unlimited source of cells for regenerative medicine. Within hematology, in vitro PSC hematopoiesis affords novel insights into blood formation and represents an exciting potential approach to generate hematopoietic and immune cell types for transplantation and transfusion. Most studies to date have focused on in vitro hematopoiesis from mouse PSCs and human PSCs. However, differences in mouse and human PSC culture protocols have complicated the translation of discoveries between these systems. We recently developed a novel chemical media formulation, expanded potential stem cell medium (EPSCM), that maintains mouse PSCs in a unique cellular state and extraembryonic differentiation capacity. Herein, we describe how EPSCM can be directly used to stably maintain human PSCs. We further demonstrate that human PSCs maintained in EPSCM can spontaneously form embryoid bodies and undergo in vitro hematopoiesis using a simple differentiation protocol, similar to mouse PSC differentiation. EPSCM-maintained human PSCs generated at least two hematopoietic cell populations, which displayed distinct transcriptional profiles by RNA-sequencing (RNA-seq) analysis. EPSCM also supports gene targeting using homologous recombination, affording generation of an SPI1 (PU.1) reporter PSC line to study and track in vitro hematopoiesis. EPSCM therefore provides a useful tool not only to study pluripotency but also hematopoietic cell specification and developmental-lineage commitment.


Asunto(s)
Medios de Cultivo/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula/métodos , Ciclo Celular , Linaje de la Célula , Células Cultivadas , Técnicas de Reprogramación Celular , Cuerpos Embrioides/efectos de los fármacos , Fibroblastos/citología , Genes Reporteros , Células Madre Embrionarias Humanas/citología , Humanos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/trasplante , Análisis de Secuencia de ARN , Especificidad de la Especie , Trasplante de Células Madre/efectos adversos , Teratoma/etiología
3.
Sci Rep ; 8(1): 15374, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30337647

RESUMEN

ß-cell replacement has been proposed as an effective treatment for some forms of diabetes, and in vitro methods for ß-cell generation are being extensively explored. A potential source of ß-cells comes from fate conversion of exocrine pancreatic cells into the endocrine lineage, by overexpression of three regulators of pancreatic endocrine formation and ß-cell identity, Ngn3, Pdx1 and MafA. Pancreatic ductal organoid cultures have recently been developed that can be expanded indefinitely, while maintaining the potential to differentiate into the endocrine lineage. Here, using mouse pancreatic ductal organoids, we see that co-expression of Ngn3, Pdx1 and MafA are required and sufficient to generate cells that express insulin and resemble ß-cells transcriptome-wide. Efficiency of ß-like cell generation can be significantly enhanced by preventing phosphorylation of Ngn3 protein and further augmented by conditions promoting differentiation. Taken together, our new findings underline the potential of ductal organoid cultures as a source material for generation of ß-like cells and demonstrate that post-translational regulation of reprogramming factors can be exploited to enhance ß-cell generation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Reprogramación Celular , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Organoides/metabolismo , Conductos Pancreáticos/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células HEK293 , Humanos , Células Secretoras de Insulina/citología , Ratones , Proteínas del Tejido Nervioso/genética , Organoides/citología , Conductos Pancreáticos/citología , Fosforilación
4.
Dev Cell ; 46(3): 360-375.e5, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30057275

RESUMEN

Pancreas development involves a coordinated process in which an early phase of cell segregation is followed by a longer phase of lineage restriction, expansion, and tissue remodeling. By combining clonal tracing and whole-mount reconstruction with proliferation kinetics and single-cell transcriptional profiling, we define the functional basis of pancreas morphogenesis. We show that the large-scale organization of mouse pancreas can be traced to the activity of self-renewing precursors positioned at the termini of growing ducts, which act collectively to drive serial rounds of stochastic ductal bifurcation balanced by termination. During this phase of branching morphogenesis, multipotent precursors become progressively fate-restricted, giving rise to self-renewing acinar-committed precursors that are conveyed with growing ducts, as well as ductal progenitors that expand the trailing ducts and give rise to delaminating endocrine cells. These findings define quantitatively how the functional behavior and lineage progression of precursor pools determine the large-scale patterning of pancreatic sub-compartments.


Asunto(s)
Linaje de la Célula , Células Endocrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Organogénesis/fisiología , Páncreas/crecimiento & desarrollo , Células Acinares/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Morfogénesis/fisiología , Células Madre/metabolismo
5.
Blood ; 131(21): e1-e11, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29588278

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Mutación , Proteínas Proto-Oncogénicas c-kit/deficiencia , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Transcriptoma
6.
Proc Natl Acad Sci U S A ; 114(23): 5822-5829, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28584094

RESUMEN

Adult blood contains a mixture of mature cell types, each with specialized functions. Single hematopoietic stem cells (HSCs) have been functionally shown to generate all mature cell types for the lifetime of the organism. Differentiation of HSCs toward alternative lineages must be balanced at the population level by the fate decisions made by individual cells. Transcription factors play a key role in regulating these decisions and operate within organized regulatory programs that can be modeled as transcriptional regulatory networks. As dysregulation of single HSC fate decisions is linked to fatal malignancies such as leukemia, it is important to understand how these decisions are controlled on a cell-by-cell basis. Here we developed and applied a network inference method, exploiting the ability to infer dynamic information from single-cell snapshot expression data based on expression profiles of 48 genes in 2,167 blood stem and progenitor cells. This approach allowed us to infer transcriptional regulatory network models that recapitulated differentiation of HSCs into progenitor cell types, focusing on trajectories toward megakaryocyte-erythrocyte progenitors and lymphoid-primed multipotent progenitors. By comparing these two models, we identified and subsequently experimentally validated a difference in the regulation of nuclear factor, erythroid 2 (Nfe2) and core-binding factor, runt domain, alpha subunit 2, translocated to, 3 homolog (Cbfa2t3h) by the transcription factor Gata2. Our approach confirms known aspects of hematopoiesis, provides hypotheses about regulation of HSC differentiation, and is widely applicable to other hierarchical biological systems to uncover regulatory relationships.


Asunto(s)
Redes Reguladoras de Genes , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Algoritmos , Animales , Diferenciación Celular , Ratones Endogámicos C57BL , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
7.
Dev Cell ; 41(3): 274-286.e5, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28457793

RESUMEN

The proneural transcription factor Neurogenin3 (Ngn3) plays a critical role in pancreatic endocrine cell differentiation, although regulation of Ngn3 protein is largely unexplored. Here we demonstrate that Ngn3 protein undergoes cyclin-dependent kinase (Cdk)-mediated phosphorylation on multiple serine-proline sites. Replacing wild-type protein with a phosphomutant form of Ngn3 increases α cell generation, the earliest endocrine cell type to be formed in the developing pancreas. Moreover, un(der)phosphorylated Ngn3 maintains insulin expression in adult ß cells in the presence of elevated c-Myc and enhances endocrine specification during ductal reprogramming. Mechanistically, preventing multi-site phosphorylation enhances both Ngn3 stability and DNA binding, promoting the increased expression of target genes that drive differentiation. Therefore, multi-site phosphorylation of Ngn3 controls its ability to promote pancreatic endocrine differentiation and to maintain ß cell function in the presence of pro-proliferation cues and could be manipulated to promote and maintain endocrine differentiation in vitro and in vivo.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Proteínas del Tejido Nervioso/metabolismo , Animales , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Páncreas/metabolismo , Fosforilación , Transducción de Señal/fisiología
8.
Blood ; 128(8): e20-31, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27365425

RESUMEN

Maintenance of the blood system requires balanced cell fate decisions by hematopoietic stem and progenitor cells (HSPCs). Because cell fate choices are executed at the individual cell level, new single-cell profiling technologies offer exciting possibilities for mapping the dynamic molecular changes underlying HSPC differentiation. Here, we have used single-cell RNA sequencing to profile more than 1600 single HSPCs, and deep sequencing has enabled detection of an average of 6558 protein-coding genes per cell. Index sorting, in combination with broad sorting gates, allowed us to retrospectively assign cells to 12 commonly sorted HSPC phenotypes while also capturing intermediate cells typically excluded by conventional gating. We further show that independently generated single-cell data sets can be projected onto the single-cell resolution expression map to directly compare data from multiple groups and to build and refine new hypotheses. Reconstruction of differentiation trajectories reveals dynamic expression changes associated with early lymphoid, erythroid, and granulocyte-macrophage differentiation. The latter two trajectories were characterized by common upregulation of cell cycle and oxidative phosphorylation transcriptional programs. By using external spike-in controls, we estimate absolute messenger RNA (mRNA) levels per cell, showing for the first time that despite a general reduction in total mRNA, a subset of genes shows higher expression levels in immature stem cells consistent with active maintenance of the stem-cell state. Finally, we report the development of an intuitive Web interface as a new community resource to permit visualization of gene expression in HSPCs at single-cell resolution for any gene of choice.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , Análisis de la Célula Individual/métodos , Animales , Biomarcadores/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética
9.
FEBS Lett ; 590(22): 4052-4067, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27259698

RESUMEN

Haematopoietic stem and progenitor cells (HSPCs) sit at the top of the haematopoietic hierarchy, and their fate choices need to be carefully controlled to ensure balanced production of all mature blood cell types. As cell fate decisions are made at the level of the individual cells, recent technological advances in measuring gene and protein expression in increasingly large numbers of single cells have been rapidly adopted to study both normal and pathological HSPC function. In this review we emphasise the importance of combining the correct computational models with single-cell experimental techniques, and illustrate how such integrated approaches have been used to resolve heterogeneities in populations, reconstruct lineage differentiation, identify regulatory relationships and link molecular profiling to cellular function.


Asunto(s)
Diferenciación Celular/genética , Células Madre Hematopoyéticas/citología , Análisis de la Célula Individual , Animales , Linaje de la Célula/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Células Madre Hematopoyéticas/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...