Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 57(14): 1742-1757.e5, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35803279

RESUMEN

Alveolar epithelial cell fate decisions drive lung development and regeneration. Using transcriptomic and epigenetic profiling coupled with genetic mouse and organoid models, we identified the transcription factor Klf5 as an essential determinant of alveolar epithelial cell fate across the lifespan. We show that although dispensable for both adult alveolar epithelial type 1 (AT1) and alveolar epithelial type 2 (AT2) cell homeostasis, Klf5 enforces AT1 cell lineage fidelity during development. Using infectious and non-infectious models of acute respiratory distress syndrome, we demonstrate that Klf5 represses AT2 cell proliferation and enhances AT2-AT1 cell differentiation in a spatially restricted manner during lung regeneration. Moreover, ex vivo organoid assays identify that Klf5 reduces AT2 cell sensitivity to inflammatory signaling to drive AT2-AT1 cell differentiation. These data define the roll of a major transcriptional regulator of AT1 cell lineage commitment and of the AT2 cell response to inflammatory crosstalk during lung regeneration.


Asunto(s)
Células Epiteliales Alveolares , Pulmón , Células Epiteliales Alveolares/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Organogénesis , Factores de Transcripción/metabolismo
2.
Nature ; 604(7904): 120-126, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355013

RESUMEN

The human lung differs substantially from its mouse counterpart, resulting in a distinct distal airway architecture affected by disease pathology in chronic obstructive pulmonary disease. In humans, the distal branches of the airway interweave with the alveolar gas-exchange niche, forming an anatomical structure known as the respiratory bronchioles. Owing to the lack of a counterpart in mouse, the cellular and molecular mechanisms that govern respiratory bronchioles in the human lung remain uncharacterized. Here we show that human respiratory bronchioles contain a unique secretory cell population that is distinct from cells in larger proximal airways. Organoid modelling reveals that these respiratory airway secretory (RAS) cells act as unidirectional progenitors for alveolar type 2 cells, which are essential for maintaining and regenerating the alveolar niche. RAS cell lineage differentiation into alveolar type 2 cells is regulated by Notch and Wnt signalling. In chronic obstructive pulmonary disease, RAS cells are altered transcriptionally, corresponding to abnormal alveolar type 2 cell states, which are associated with smoking exposure in both humans and ferrets. These data identify a distinct progenitor in a region of the human lung that is not found in mouse that has a critical role in maintaining the gas-exchange compartment and is altered in chronic lung disease.


Asunto(s)
Bronquiolos , Hurones , Células Madre Multipotentes , Alveolos Pulmonares , Animales , Bronquiolos/citología , Linaje de la Célula , Humanos , Pulmón/patología , Ratones , Células Madre Multipotentes/citología , Alveolos Pulmonares/citología , Enfermedad Pulmonar Obstructiva Crónica
3.
Cell Rep ; 35(6): 109092, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979629

RESUMEN

Alveolar epithelial type 2 (AT2) cells integrate signals from multiple molecular pathways to proliferate and differentiate to drive regeneration of the lung alveolus. Utilizing in vivo genetic and ex vivo organoid models, we investigated the role of Fgfr2 signaling in AT2 cells across the lifespan and during adult regeneration after influenza infection. We show that, although dispensable for adult homeostasis, Fgfr2 restricts AT2 cell fate during postnatal lung development. Using an unbiased computational imaging approach, we demonstrate that Fgfr2 promotes AT2 cell proliferation and restrains differentiation in actively regenerating areas after injury. Organoid assays reveal that Fgfr2-deficient AT2 cells remain competent to respond to multiple parallel proliferative inputs. Moreover, genetic blockade of AT2 cell cytokinesis demonstrates that cell division and differentiation are uncoupled during alveolar regeneration. These data reveal that Fgfr2 maintains AT2 cell fate, balancing proliferation and differentiation during lung alveolar regeneration.


Asunto(s)
Lesión Pulmonar Aguda/fisiopatología , Células Epiteliales Alveolares/metabolismo , Pulmón/patología , Animales , Proliferación Celular , Humanos , Ratones
4.
Cell Stem Cell ; 28(10): 1775-1789.e5, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33974915

RESUMEN

Regeneration of the architecturally complex alveolar niche of the lung requires precise temporal and spatial control of epithelial cell behavior. Injury can lead to a permanent reduction in gas exchange surface area and respiratory function. Using mouse models, we show that alveolar type 1 (AT1) cell plasticity is a major and unappreciated mechanism that drives regeneration, beginning in the early postnatal period during alveolar maturation. Upon acute neonatal lung injury, AT1 cells reprogram into alveolar type 2 (AT2) cells, promoting alveolar regeneration. In contrast, the ability of AT2 cells to regenerate AT1 cells is restricted to the mature lung. Unbiased genomic assessment reveals that this previously unappreciated level of plasticity is governed by the preferential activity of Hippo signaling in the AT1 cell lineage. Thus, cellular plasticity is a temporally acquired trait of the alveolar epithelium and presents an alternative mode of tissue regeneration in the postnatal lung.


Asunto(s)
Células Epiteliales Alveolares , Pulmón , Animales , Homeostasis , Ratones , Mucosa Respiratoria , Transducción de Señal
5.
Science ; 371(6534)2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33707239

RESUMEN

The lung alveolus is the functional unit of the respiratory system required for gas exchange. During the transition to air breathing at birth, biophysical forces are thought to shape the emerging tissue niche. However, the intercellular signaling that drives these processes remains poorly understood. Applying a multimodal approach, we identified alveolar type 1 (AT1) epithelial cells as a distinct signaling hub. Lineage tracing demonstrates that AT1 progenitors align with receptive, force-exerting myofibroblasts in a spatial and temporal manner. Through single-cell chromatin accessibility and pathway expression (SCAPE) analysis, we demonstrate that AT1-restricted ligands are required for myofibroblasts and alveolar formation. These studies show that the alignment of cell fates, mediated by biophysical and AT1-derived paracrine signals, drives the extensive tissue remodeling required for postnatal respiration.


Asunto(s)
Linaje de la Célula/genética , Epigénesis Genética , Alveolos Pulmonares/embriología , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Animales , Células Cultivadas , Señales (Psicología) , Epigenómica , Humanos , Ratones , Ratones Transgénicos , Miofibroblastos/citología , Miofibroblastos/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , RNA-Seq/métodos , Transducción de Señal , Análisis de la Célula Individual , Transcriptoma
6.
Nat Cell Biol ; 22(10): 1197-1210, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32989251

RESUMEN

Alveolar epithelial regeneration is essential for recovery from devastating lung diseases. This process occurs when type II alveolar pneumocytes (AT2 cells) proliferate and transdifferentiate into type I alveolar pneumocytes (AT1 cells). We used genome-wide analysis of chromatin accessibility and gene expression following acute lung injury to elucidate repair mechanisms. AT2 chromatin accessibility changed substantially following injury to reveal STAT3 binding motifs adjacent to genes that regulate essential regenerative pathways. Single-cell transcriptome analysis identified brain-derived neurotrophic factor (Bdnf) as a STAT3 target gene with newly accessible chromatin in a unique population of regenerating AT2 cells. Furthermore, the BDNF receptor tropomyosin receptor kinase B (TrkB) was enriched on mesenchymal alveolar niche cells (MANCs). Loss or blockade of AT2-specific Stat3, Bdnf or mesenchyme-specific TrkB compromised repair and reduced Fgf7 expression by niche cells. A TrkB agonist improved outcomes in vivo following lung injury. These data highlight the biological and therapeutic importance of the STAT3-BDNF-TrkB axis in orchestrating alveolar epithelial regeneration.


Asunto(s)
Células Epiteliales Alveolares/citología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Lesión Pulmonar/prevención & control , Glicoproteínas de Membrana/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptor trkB/metabolismo , Regeneración , Factor de Transcripción STAT3/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Humanos , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Masculino , Glicoproteínas de Membrana/genética , Proteínas Tirosina Quinasas/genética , Receptor trkB/genética , Factor de Transcripción STAT3/genética
7.
J Cell Sci ; 133(10)2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32317397

RESUMEN

A large fraction of epigenetically silent heterochromatin is anchored to the nuclear periphery via 'tethering proteins' that function to bridge heterochromatin and the nuclear membrane or nuclear lamina. We previously identified a human tethering protein, PRR14, that binds heterochromatin through an N-terminal domain, but the mechanism and regulation of nuclear lamina association remained to be investigated. Here we identify an evolutionarily conserved PRR14 nuclear lamina binding domain (LBD) that is both necessary and sufficient for positioning of PRR14 at the nuclear lamina. We show that PRR14 associates dynamically with the nuclear lamina, and provide evidence that such dynamics are regulated through phosphorylation and dephosphorylation of the LBD. Furthermore, we identify a PP2A phosphatase recognition motif within the evolutionarily conserved C-terminal Tantalus domain of PRR14. Disruption of this motif affects PRR14 localization to the nuclear lamina. The overall findings demonstrate a heterochromatin anchoring mechanism whereby the PRR14 tether simultaneously binds heterochromatin and the nuclear lamina through two separable modular domains. Our findings also describe an optimal PRR14 LBD fragment that could be used for efficient targeting of fusion proteins to the nuclear lamina.


Asunto(s)
Heterocromatina , Lámina Nuclear , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Lámina Nuclear/genética , Lámina Nuclear/metabolismo , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA