Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Biochem Biophys Res Commun ; 629: 101-105, 2022 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-36116371

RESUMEN

Accumulated studies have suggested that bone morphogenetic proteins (BMPs) are critical for skin development. However, it remains elusive how BMP signaling via ALK2 (aka ACVR1), one of the important BMP type I receptors, regulates keratinocyte differentiation. To address this question, we utilized a genetic system that enhances BMP signaling via ALK2 in an epidermis-specific manner in mice (hereafter ca-Alk2:K14-Cre). Ca-Alk2:K14-Cre mice displayed a sticky and hairless skin phenotype with a thinner epidermis incapable of differentiating. Although cellular proliferation and survival were comparable between wild-type and ca-Alk2:K14-Cre mice, skin differentiation was severely hampered in ca-Alk2:K14-Cre mice. To uncover the mechanism of altered keratinocyte differentiation, we performed a transcriptome analysis. As a result, we found that the expression levels of cell cycle inhibitor p21 were increased in ca-Alk2:K14-Cre mice. Our findings suggest that aberrant BMP signaling via ALK2 positively regulates p21 expression that attenuates keratinocyte differentiation, and further highlights the critical role of BMP signaling in skin development.


Asunto(s)
Receptores de Activinas Tipo I , Proteínas Morfogenéticas Óseas , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/genética , Queratinocitos/metabolismo , Ratones , Transducción de Señal/genética
2.
Proc Natl Acad Sci U S A ; 116(41): 20545-20555, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31548395

RESUMEN

The extraordinarily thin alveolar type 1 (AT1) cell constitutes nearly the entire gas exchange surface and allows passive diffusion of oxygen into the blood stream. Despite such an essential role, the transcriptional network controlling AT1 cells remains unclear. Using cell-specific knockout mouse models, genomic profiling, and 3D imaging, we found that NK homeobox 2-1 (Nkx2-1) is expressed in AT1 cells and is required for the development and maintenance of AT1 cells. Without Nkx2-1, developing AT1 cells lose 3 defining features-molecular markers, expansive morphology, and cellular quiescence-leading to alveolar simplification and lethality. NKX2-1 is also cell-autonomously required for the same 3 defining features in mature AT1 cells. Intriguingly, Nkx2-1 mutant AT1 cells activate gastrointestinal (GI) genes and form dense microvilli-like structures apically. Single-cell RNA-seq supports a linear transformation of Nkx2-1 mutant AT1 cells toward a GI fate. Whole lung ChIP-seq shows NKX2-1 binding to 68% of genes that are down-regulated upon Nkx2-1 deletion, including 93% of known AT1 genes, but near-background binding to up-regulated genes. Our results place NKX2-1 at the top of the AT1 cell transcriptional hierarchy and demonstrate remarkable plasticity of an otherwise terminally differentiated cell type.


Asunto(s)
Células Epiteliales Alveolares/citología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Pulmón/crecimiento & desarrollo , Mutación , Organogénesis , Factor Nuclear Tiroideo 1/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Diferenciación Celular , Pulmón/metabolismo , Ratones , Análisis de la Célula Individual , Factor Nuclear Tiroideo 1/antagonistas & inhibidores , Factor Nuclear Tiroideo 1/genética
3.
Development ; 145(5)2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29440304

RESUMEN

The entire lung epithelium arises from SRY box 9 (SOX9)-expressing progenitors that form the respiratory tree and differentiate into airway and alveolar cells. Despite progress in understanding their initial specification within the embryonic foregut, how these progenitors are subsequently maintained is less clear. Using inducible, progenitor-specific genetic mosaic mouse models, we showed that ß-catenin (CTNNB1) maintains lung progenitors by promoting a hierarchical lung progenitor gene signature, suppressing gastrointestinal (GI) genes, and regulating NK2 homeobox 1 (NKX2.1) and SRY box 2 (SOX2) in a developmental stage-dependent manner. At the early, but not later, stage post-lung specification, CTNNB1 cell-autonomously maintained normal NKX2.1 expression levels and suppressed ectopic SOX2 expression. Genetic epistasis analyses revealed that CTNNB1 is required for fibroblast growth factor (Fgf)/Kirsten rat sarcoma viral oncogene homolog (Kras)-mediated promotion of the progenitors. In silico screening of Eurexpress and translating ribosome affinity purification (TRAP)-RNAseq identified a progenitor gene signature, a subset of which depends on CTNNB1. Wnt signaling also maintained NKX2.1 expression and suppressed GI genes in cultured human lung progenitors derived from embryonic stem cells.


Asunto(s)
Linaje de la Célula/genética , Células Madre Embrionarias/metabolismo , Células Epiteliales/citología , Pulmón/embriología , Mucosa Respiratoria/citología , Mucosa Respiratoria/embriología , beta Catenina/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Ratones Noqueados , Embarazo , Mucosa Respiratoria/metabolismo , Transcriptoma , beta Catenina/genética
4.
FASEB J ; 28(7): 2852-63, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24627544

RESUMEN

Arginine, the common substrate for production of nitric oxide (NO) and polyamines in mammals, increases in the uterine lumen during the peri-implantation period of pregnancy. However, functional roles of arginine within the uterine lumen for conceptus (embryo and extraembryonic membranes) development have not been elucidated in vivo. To assess roles of arginine in reproductive tissue for survival and development of the conceptus, we conducted an in vivo morpholino antisense oligonucleotide (MAO)-mediated knockdown of SLC7A1 mRNA, the arginine transporter in ovine conceptus trophectoderm (Tr). Translational knockdown of SLC7A1 mRNA resulted in retarded conceptus development and abnormal function compared to MAO control. Use of MAO-SLC7A1 knockdown in conceptuses decreased arginine transport (73%, P<0.01), the abundance of ornithine decarboxylase, and nitric oxide synthase (NOS3) proteins, arginine-related amino acids [citrulline (76%, P<0.05) and ornithine (40%, P<0.05)], and polyamines, which likely accounts for their retarded development. Also, no alternative arginine precursors (glutamine and glutamate), isoforms of nitric oxide synthase (NOS1 and NOS2), or alternative pathways for polyamine biosynthesis via arginine decarboxylase and agmatinase were activated to rescue conceptus development. Collectively, SLC7A1 is the key transporter of arginine by conceptus Tr, and arginine is essential for conceptus survival and development.-Wang, X., Frank, J. W., Little, D. R., Dunlap, K. A., Satterfield, M. C., Burghardt, R. C., Hansen, T. R., Wu, G., and Bazer, F. W. Functional role of arginine during the peri-implantation period of pregnancy. I. Consequences of loss of function of arginine transporter SLC7A1 mRNA in ovine conceptus trophectoderm.


Asunto(s)
Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/genética , Transportador de Aminoácidos Catiónicos 1/metabolismo , Implantación del Embrión/fisiología , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Arginina/genética , Implantación del Embrión/genética , Endometrio/metabolismo , Endometrio/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ornitina Descarboxilasa/genética , Ornitina Descarboxilasa/metabolismo , Poliaminas/metabolismo , Embarazo , ARN Mensajero/genética , Ovinos/genética , Ovinos/metabolismo , Útero/metabolismo , Útero/fisiología
5.
Nat Commun ; 12(1): 2509, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33947861

RESUMEN

Differential transcription of identical DNA sequences leads to distinct tissue lineages and then multiple cell types within a lineage, an epigenetic process central to progenitor and stem cell biology. The associated genome-wide changes, especially in native tissues, remain insufficiently understood, and are hereby addressed in the mouse lung, where the same lineage transcription factor NKX2-1 promotes the diametrically opposed alveolar type 1 (AT1) and AT2 cell fates. Here, we report that the cell-type-specific function of NKX2-1 is attributed to its differential chromatin binding that is acquired or retained during development in coordination with partner transcriptional factors. Loss of YAP/TAZ redirects NKX2-1 from its AT1-specific to AT2-specific binding sites, leading to transcriptionally exaggerated AT2 cells when deleted in progenitors or AT1-to-AT2 conversion when deleted after fate commitment. Nkx2-1 mutant AT1 and AT2 cells gain distinct chromatin accessible sites, including those specific to the opposite fate while adopting a gastrointestinal fate, suggesting an epigenetic plasticity unexpected from transcriptional changes. Our genomic analysis of single or purified cells, coupled with precision genetics, provides an epigenetic basis for alveolar cell fate and potential, and introduces an experimental benchmark for deciphering the in vivo function of lineage transcription factors.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Epigénesis Genética , Histonas/metabolismo , Células Madre/metabolismo , Factor Nuclear Tiroideo 1/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Epiteliales Alveolares/citología , Animales , Linaje de la Célula , Inmunoprecipitación de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Pulmón/metabolismo , Ratones , Mutación , Unión Proteica , RNA-Seq , Análisis de la Célula Individual , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
6.
Cancer Discov ; 11(10): 2506-2523, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33972311

RESUMEN

Little is known of the geospatial architecture of individual cell populations in lung adenocarcinoma (LUAD) evolution. Here, we perform single-cell RNA sequencing of 186,916 cells from five early-stage LUADs and 14 multiregion normal lung tissues of defined spatial proximities from the tumors. We show that cellular lineages, states, and transcriptomic features geospatially evolve across normal regions to LUADs. LUADs also exhibit pronounced intratumor cell heterogeneity within single sites and transcriptional lineage-plasticity programs. T regulatory cell phenotypes are increased in normal tissues with proximity to LUAD, in contrast to diminished signatures and fractions of cytotoxic CD8+ T cells, antigen-presenting macrophages, and inflammatory dendritic cells. We further find that the LUAD ligand-receptor interactome harbors increased expression of epithelial CD24, which mediates protumor phenotypes. These data provide a spatial atlas of LUAD evolution, and a resource for identification of targets for its treatment. SIGNIFICANCE: The geospatial ecosystem of the peripheral lung and early-stage LUAD is not known. Our multiregion single-cell sequencing analyses unravel cell populations, states, and phenotypes in the spatial and ecologic evolution of LUAD from the lung that comprise high-potential targets for early interception.This article is highlighted in the In This Issue feature, p. 2355.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Linfocitos T CD8-positivos , Neoplasias Pulmonares/patología , Microambiente Tumoral , Humanos , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA