Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 417
Filtrar
1.
Elife ; 102021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34586063

RESUMEN

Lung epithelial progenitors differentiate into alveolar type 1 (AT1) and type 2 (AT2) cells. These cells form the air-blood interface and secrete surfactant, respectively, and are essential for lung maturation and function. Current protocols to derive and culture alveolar cells do not faithfully recapitulate the architecture of the distal lung, which influences cell fate patterns in vivo. Here, we report serum-free conditions that allow for growth and differentiation of mouse distal lung epithelial progenitors. We find that Collagen I promotes the differentiation of flattened, polarized AT1 cells. Using these organoids, we performed a chemical screen to investigate WNT signaling in epithelial differentiation. We identify an association between Casein Kinase activity and maintenance of an AT2 expression signature; Casein Kinase inhibition leads to an increase in AT1/progenitor cell ratio. These organoids provide a simplified model of alveolar differentiation and constitute a scalable screening platform to identify and analyze cell differentiation mechanisms.


Asunto(s)
Diferenciación Celular , Alveolos Pulmonares/citología , Células Madre/citología , Animales , Caseína Quinasas/antagonistas & inhibidores , Caseína Quinasas/metabolismo , Células Cultivadas , Colágeno Tipo I/metabolismo , Medio de Cultivo Libre de Suero , Células Epiteliales/citología , Células Epiteliales/metabolismo , Marcadores Genéticos , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/embriología , Alveolos Pulmonares/enzimología , Alveolos Pulmonares/metabolismo , Transcripción Genética , Vía de Señalización Wnt
2.
Cell Death Dis ; 12(9): 841, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34497269

RESUMEN

Although aberrant alveolar myofibroblasts (AMYFs) proliferation and differentiation are often associated with abnormal lung development and diseases, such as bronchopulmonary dysplasia (BPD), chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF), epigenetic mechanisms regulating proliferation and differentiation of AMYFs remain poorly understood. Protein arginine methyltransferase 7 (PRMT7) is the only reported type III enzyme responsible for monomethylation of arginine residue on both histone and nonhistone substrates. Here we provide evidence for PRMT7's function in regulating AMYFs proliferation and differentiation during lung alveologenesis. In PRMT7-deficient mice, we found reduced AMYFs proliferation and differentiation, abnormal elastin deposition, and failure of alveolar septum formation. We further shown that oncogene forkhead box M1 (Foxm1) is a direct target of PRMT7 and that PRMT7-catalyzed monomethylation at histone H4 arginine 3 (H4R3me1) directly associate with chromatin of Foxm1 to activate its transcription, and thereby regulate of cell cycle-related genes to inhibit AMYFs proliferation and differentiation. Overexpression of Foxm1 in isolated myofibroblasts (MYFs) significantly rescued PRMT7-deficiency-induced cell proliferation and differentiation defects. Thus, our results reveal a novel epigenetic mechanism through which PRMT7-mediated histone arginine monomethylation activates Foxm1 transcriptional expression to regulate AMYFs proliferation and differentiation during lung alveologenesis and may represent a potential target for intervention in pulmonary diseases.


Asunto(s)
Diferenciación Celular , Proteína Forkhead Box M1/metabolismo , Miofibroblastos/citología , Miofibroblastos/metabolismo , Organogénesis , Proteína-Arginina N-Metiltransferasas/metabolismo , Alveolos Pulmonares/embriología , Actinas/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Proliferación Celular/genética , Elastina/metabolismo , Epigénesis Genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Antígeno Ki-67/metabolismo , Mesodermo/embriología , Ratones , Modelos Biológicos , Especificidad de Órganos , Organogénesis/genética , Fenotipo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteína-Arginina N-Metiltransferasas/deficiencia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L814-L826, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431413

RESUMEN

Accurate fluid pressure in the fetal lung is critical for its development, especially at the beginning of the saccular stage when alveolar epithelial type 1 (AT1) and type 2 (AT2) cells differentiate from the epithelial progenitors. Despite our growing understanding of the role of physical forces in lung development, the molecular mechanisms that regulate the transduction of mechanical stretch to alveolar differentiation remain elusive. To simulate lung distension, we optimized both an ex vivo model with precision cut lung slices and an in vivo model of fetal tracheal occlusion. Increased mechanical tension showed to improve alveolar maturation and differentiation toward AT1. By manipulating ROCK pathway, we demonstrate that stretch-induced Yap/Taz activation promotes alveolar differentiation toward AT1 phenotype via ROCK activity. Our findings show that balanced ROCK-Yap/Taz signaling is essential to regulate AT1 differentiation in response to mechanical stretching of the fetal lung, which might be helpful in improving lung development and regeneration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Epiteliales Alveolares/fisiología , Mecanotransducción Celular/fisiología , Alveolos Pulmonares/embriología , Quinasas Asociadas a rho/metabolismo , Células Epiteliales Alveolares/citología , Animales , Recuento de Células , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Ratones , Microscopía Electrónica de Rastreo , Organogénesis/fisiología , Transducción de Señal/fisiología , Proteínas Señalizadoras YAP
4.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33830085

RESUMEN

The molecular mechanisms by which endothelial cells (ECs) regulate pulmonary vascularization and contribute to alveolar epithelial cell development during lung morphogenesis remain unknown. We tested the hypothesis that delta-like 4 (DLL4), an EC Notch ligand, is critical for alveolarization by combining lung mapping and functional studies in human tissue and DLL4-haploinsufficient mice (Dll4+/lacz). DLL4 expressed in a PECAM-restricted manner in capillaries, arteries, and the alveolar septum from the canalicular to alveolar stage in mice and humans. Dll4 haploinsufficiency resulted in exuberant, nondirectional vascular patterning at E17.5 and P6, followed by smaller capillaries and fewer intermediate blood vessels at P14. Vascular defects coincided with polarization of lung EC expression toward JAG1-NICD-HES1 signature and decreased tip cell-like (Car4) markers. Dll4+/lacZ mice had impaired terminal bronchiole development at the canalicular stage and impaired alveolarization upon lung maturity. We discovered that alveolar type I cell (Aqp5) markers progressively decreased in Dll4+/lacZ mice after birth. Moreover, in human lung EC, DLL4 deficiency programmed a hypersprouting angiogenic phenotype cell autonomously. In conclusion, DLL4 is expressed from the canalicular to alveolar stage in mice and humans, and Dll4 haploinsufficiency programs dysmorphic microvascularization, impairing alveolarization. Our study reveals an obligate role for DLL4-regulated angiogenesis in distal lung morphogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/metabolismo , Pulmón/irrigación sanguínea , Pulmón/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Células Epiteliales Alveolares/fisiología , Animales , Proteínas de Unión al Calcio/genética , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia , Humanos , Hipoxia , Ratones Endogámicos C57BL , Ratones Mutantes , Neovascularización Fisiológica/genética , Alveolos Pulmonares/citología , Alveolos Pulmonares/embriología , Alveolos Pulmonares/metabolismo
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.
Dev Dyn ; 250(4): 482-496, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33169483

RESUMEN

Lung alveologenesis, formation of the alveolar region, allows sufficient gas exchange surface to be packed inside the chest cavity yet with orderly connection to the trachea. The real-life alveolar region, however, bears little resemblance to idealized cartoons owing to its three-dimensional nature, nonuniform shape, and mostly air-filled void. This morphological complexity is matched by its cellular complexity-comprised of intermixed and often tangled cells of the epithelial, mesenchymal, endothelial, and immune lineages. Modern imaging, genetics, and genomics are shedding light on and updating traditional views of alveologenesis. Accordingly, this review describes a cell-centric 3-phase definition of alveologenesis and discusses its failure in diseases and possible reactivation during regeneration.


Asunto(s)
Alveolos Pulmonares/citología , Alveolos Pulmonares/embriología , Animales , Humanos , Organogénesis , Alveolos Pulmonares/fisiología , Regeneración
7.
Elife ; 92020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32394892

RESUMEN

Alveolar formation increases the surface area for gas-exchange and is key to the physiological function of the lung. Alveolar epithelial cells, myofibroblasts and endothelial cells undergo coordinated morphogenesis to generate epithelial folds (secondary septa) to form alveoli. A mechanistic understanding of alveologenesis remains incomplete. We found that the planar cell polarity (PCP) pathway is required in alveolar epithelial cells and myofibroblasts for alveologenesis in mammals. Our studies uncovered a Wnt5a-Ror2-Vangl2 cascade that endows cellular properties and novel mechanisms of alveologenesis. This includes PDGF secretion from alveolar type I and type II cells, cell shape changes of type I cells and migration of myofibroblasts. All these cellular properties are conferred by changes in the cytoskeleton and represent a new facet of PCP function. These results extend our current model of PCP signaling from polarizing a field of epithelial cells to conferring new properties at subcellular levels to regulate collective cell behavior.


The lungs enable the exchange of gases between inhaled air and the bloodstream. This exchange happens in structures called alveoli, which have a large surface area that aids in efficient gas exchange. Shortly after birth in mice, or during the last few months before birth in humans, alveoli develop folds called secondary septa that increase their surface area and improve the efficiency of gas exchange. Several types of cells work together to form secondary septa. Surface cells called epithelia and underlying "myofibroblast" cells and small blood vessels must both communicate and move together to build the septa. The processes that control the formation of septa have not been fully studied. In other cases, a cell signaling pathway known as the planar cell polarity (PCP) pathway has been shown to help coordinate cell movements. The PCP pathway works by changing the cytoskeleton of cells, which is the series of protein fibers that give cells their shape and structure and the ability to move. Zhang et al. have now studied septa in mouse lungs and revealed how three genes ­ Wnt5a, Ror2 and Vangl2 ­ in the PCP pathway control this process. This pathway oversees changes to the cytoskeleton in both epithelial cells and myofibroblasts, helping the cells to change shape and move together to form septa. Unusually, the PCP pathway has different effects in different cells, rather than affecting all cells similarly. This is partly due to so-called PDGF signals from the epithelial cells that help to guide the growth and movement of myofibroblasts. This process is helped by the epithelial cells changing their shape to accommodate myofibroblasts during septa formation. Further analysis also showed reduced PCP signaling in patients with chronic obstructive pulmonary disease, also known as COPD. This could be a factor in the extensive lung damage seen in these patients. These findings help to explain a key lung development process and may provide new insights to understand lung diseases such as COPD.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Citoesqueleto/fisiología , Proteínas del Tejido Nervioso/metabolismo , Alveolos Pulmonares/fisiología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Proteína Wnt-5a/metabolismo , Actomiosina , Células Epiteliales Alveolares/citología , Animales , Polaridad Celular , Forma de la Célula , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Ligandos , Pulmón/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Morfogénesis , Miofibroblastos/citología , Miofibroblastos/fisiología , Organogénesis , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/embriología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transducción de Señal
8.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1165-L1171, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292070

RESUMEN

Bronchopulmonary dysplasia (BPD), a long-term respiratory morbidity of prematurity, is characterized by attenuated alveolar and vascular development. Supplemental oxygen and immature antioxidant defenses contribute to BPD development. Our group identified thioredoxin reductase-1 (TXNRD1) as a therapeutic target to prevent BPD. The present studies evaluated the impact of the TXNRD1 inhibitor aurothioglucose (ATG) on pulmonary responses and gene expression in newborn C57BL/6 pups treated with saline or ATG (25 mg/kg ip) within 12 h of birth and exposed to room air (21% O2) or hyperoxia (>95% O2) for 72 h. Purified RNA from lung tissues was sequenced, and differential expression was evaluated. Hyperoxic exposure altered ~2,000 genes, including pathways involved in glutathione metabolism, intrinsic apoptosis signaling, and cell cycle regulation. The isolated effect of ATG treatment was limited primarily to genes that regulate angiogenesis and vascularization. In separate studies, pups were treated as described above and returned to room air until 14 days. Vascular density analyses were performed, and ANOVA indicated an independent effect of hyperoxia on vascular density and alveolar architecture at 14 days. Consistent with RNA-seq analyses, ATG significantly increased vascular density in room air, but not in hyperoxia-exposed pups. These findings provide insights into the mechanisms by which TXNRD1 inhibitors may enhance lung development.


Asunto(s)
Aire , Aurotioglucosa/farmacología , Hiperoxia/patología , Pulmón/irrigación sanguínea , Pulmón/patología , Neovascularización Fisiológica/efectos de los fármacos , Enfermedad Aguda , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/genética , ADN/biosíntesis , Glutatión/metabolismo , Pulmón/efectos de los fármacos , Pulmón/embriología , Ratones Endogámicos C57BL , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/embriología , Alveolos Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcriptoma/genética , Regulación hacia Arriba/efectos de los fármacos
9.
Cells ; 9(2)2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32046118

RESUMEN

WNT5a is a mainly "non-canonical" WNT ligand whose dysregulation is observed in lung diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and asthma. Germline deletion of Wnt5a disrupts embryonic lung development. However, the temporal-specific function of WNT5a remains unknown. In this study, we generated a conditional loss-of-function mouse model (Wnt5aCAG) and examined the specific role of Wnt5a during the saccular and alveolar phases of lung development. The lack of Wnt5a in the saccular phase blocked distal airway expansion and attenuated differentiation of endothelial and alveolar epithelial type I (AT1) cells and myofibroblasts. Postnatal Wnt5a inactivation disrupted alveologenesis, producing a phenotype resembling human bronchopulmonary dysplasia (BPD). Mutant lungs showed hypoalveolization, but endothelial and epithelial differentiation was unaffected. The major impact of Wnt5a inactivation on alveologenesis was on myofibroblast differentiation and migration, with reduced expression of key regulatory genes. These findings were validated in vitro using isolated lung fibroblasts. Conditional inactivation of the WNT5a receptors Ror1 and Ror2 in alveolar myofibroblasts recapitulated the Wnt5aCAG phenotype, demonstrating that myofibroblast defects are the major cause of arrested alveologenesis in Wnt5aCAG lungs. Finally, we show that WNT5a is reduced in human BPD lung samples, indicating the clinical relevance and potential role for WNT5a in pathogenesis of BPD.


Asunto(s)
Organogénesis , Alveolos Pulmonares/embriología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal , Proteína Wnt-5a/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Humanos , Recién Nacido , Ratones , Modelos Biológicos , Miofibroblastos/citología
10.
Development ; 147(4)2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32001436

RESUMEN

Proper organ development depends on coordinated communication between multiple cell types. Retinoic acid (RA) is an autocrine and paracrine signaling molecule essential for the development of most organs, including the lung. Despite extensive work detailing effects of RA deficiency in early lung morphogenesis, little is known about how RA regulates late gestational lung maturation. Here, we investigate the role of the RA catabolizing protein Cyp26b1 in the lung. Cyp26b1 is highly enriched in lung endothelial cells (ECs) throughout development. We find that loss of Cyp26b1 leads to reduction of alveolar type 1 cells, failure of alveolar inflation and early postnatal lethality in mouse. Furthermore, we observe expansion of distal epithelial progenitors, but no appreciable changes in proximal airways, ECs or stromal populations. Exogenous administration of RA during late gestation partially mimics these defects; however, transcriptional analyses comparing Cyp26b1-/- with RA-treated lungs reveal overlapping, but distinct, responses. These data suggest that defects observed in Cyp26b1-/- lungs are caused by both RA-dependent and RA-independent mechanisms. This work reports crucial cellular crosstalk during lung development involving Cyp26b1-expressing endothelium and identifies a novel RA modulator in lung development.


Asunto(s)
Epitelio/embriología , Pulmón/embriología , Alveolos Pulmonares/embriología , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/fisiología , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Células Endoteliales/citología , Células Epiteliales/citología , Femenino , Regulación del Desarrollo de la Expresión Génica , Riñón/embriología , Ratones , Ratones Endogámicos C57BL , Organogénesis/efectos de los fármacos , Embarazo , Preñez , Transducción de Señal , Células Madre/citología , Tretinoina/farmacología
11.
Development ; 146(15)2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31331942

RESUMEN

Postnatal alveolar formation is the most important and the least understood phase of lung development. Alveolar pathologies are prominent in neonatal and adult lung diseases. The mechanisms of alveologenesis remain largely unknown. We inactivated Pdgfra postnatally in secondary crest myofibroblasts (SCMF), a subpopulation of lung mesenchymal cells. Lack of Pdgfra arrested alveologenesis akin to bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. The transcriptome of mutant SCMF revealed 1808 altered genes encoding transcription factors, signaling and extracellular matrix molecules. Elastin mRNA was reduced, and its distribution was abnormal. Absence of Pdgfra disrupted expression of elastogenic genes, including members of the Lox, Fbn and Fbln families. Expression of EGF family members increased when Tgfb1 was repressed in mouse. Similar, but not identical, results were found in human BPD lung samples. In vitro, blocking PDGF signaling decreased elastogenic gene expression associated with increased Egf and decreased Tgfb family mRNAs. The effect was reversible by inhibiting EGF or activating TGFß signaling. These observations demonstrate the previously unappreciated postnatal role of PDGFA/PDGFRα in controlling elastogenic gene expression via a secondary tier of signaling networks composed of EGF and TGFß.


Asunto(s)
Familia de Proteínas EGF/metabolismo , Miofibroblastos/metabolismo , Alveolos Pulmonares/embriología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Displasia Broncopulmonar/patología , Proteínas de Unión al Calcio/biosíntesis , Diferenciación Celular/fisiología , Células Cultivadas , Elastina/genética , Proteínas de la Matriz Extracelular/biosíntesis , Fibrilina-1/biosíntesis , Humanos , Ratones , Ratones Noqueados , Proteína-Lisina 6-Oxidasa/biosíntesis , ARN Mensajero/genética , Factor de Crecimiento Transformador beta1/biosíntesis
12.
J Cell Biochem ; 120(10): 16876-16887, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31144392

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common and refractory disease affecting newborn children and infants with alveolar dysplasia and declined pulmonary function. Several microRNAs (miRNAs) have been found to be differentially expressed in BPD progression. This study further explores the role of miR-421 via fibroblast growth factor 10 (Fgf10) in mice with BPD. A mouse model of BPD was established through the induction of hyperoxia, in which the expression pattern of miR-421 and Fgf10 was identified. Furthermore, adenovirus-packed vectors were injected in mice to intervene miR-421 and Fgf10 expression, including miR-421 mimics or inhibitors, and si-Fgf10 to explore the role of miR-421 and Fgf10 in BPD. The target relationship between miR-421 and Fgf10 was investigated. Inflammatory response and cell apoptosis were observed in the mice, with inflammatory cytokines and apoptosis-related factors detected by applying Reverse transcription quantitative polymerase chain reaction, Western blot analysis, and enzyme-linked immunosorbent assay. Fgf10 was confirmed as a target gene of miR-421. Elevated expression of miR-421 was evident, while Fgf10 was poorly expressed in BPD. upregulation of miR-421 and silence of Fgf10 aggravated inflammatory response in lung tissue and promoted lung cell apoptosis in BPD. The aforementioned alterations could be reversed by downregulation of miR-421. Collectively, inhibition of miR-421 can assist in the development of BPD in mice BPD by upregulating Fgf10. Therefore, the present study provides a probable target for the treatment of BPD.


Asunto(s)
Apoptosis/genética , Displasia Broncopulmonar/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , MicroARNs/genética , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Silenciador del Gen , Hiperoxia/fisiopatología , Inflamación/genética , Inflamación/patología , Ratones , Ratones Endogámicos , Alveolos Pulmonares/embriología
13.
Nat Commun ; 10(1): 1178, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862802

RESUMEN

Damage to alveoli, the gas-exchanging region of the lungs, is a component of many chronic and acute lung diseases. In addition, insufficient generation of alveoli results in bronchopulmonary dysplasia, a disease of prematurity. Therefore visualising the process of alveolar development (alveologenesis) is critical for our understanding of lung homeostasis and for the development of treatments to repair and regenerate lung tissue. Here we show live alveologenesis, using long-term, time-lapse imaging of precision-cut lung slices. We reveal that during this process, epithelial cells are highly mobile and we identify specific cell behaviours that contribute to alveologenesis: cell clustering, hollowing and cell extension. Using the cytoskeleton inhibitors blebbistatin and cytochalasin D, we show that cell migration is a key driver of alveologenesis. This study reveals important novel information about lung biology and provides a new system in which to manipulate alveologenesis genetically and pharmacologically.


Asunto(s)
Movimiento Celular/fisiología , Células Epiteliales/fisiología , Organogénesis/fisiología , Alveolos Pulmonares/embriología , Actomiosina/antagonistas & inhibidores , Actomiosina/fisiología , Animales , Animales Recién Nacidos , Movimiento Celular/efectos de los fármacos , Citocalasina D/farmacología , Células Epiteliales/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Microscopía Intravital , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Modelos Animales , Organogénesis/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Imagen de Lapso de Tiempo
14.
Development ; 146(2)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651296

RESUMEN

Organ growth and tissue homeostasis rely on the proliferation and differentiation of progenitor cell populations. In the developing lung, localized Fgf10 expression maintains distal Sox9-expressing epithelial progenitors and promotes basal cell differentiation in the cartilaginous airways. Mesenchymal Fgf10 expression is induced by Wnt signaling but inhibited by Shh signaling, and epithelial Fgf10 signaling activates ß-catenin signaling. The Hippo pathway is a well-conserved signaling cascade that regulates organ size and stem/progenitor cell behavior. Here, we show that Hippo signaling promotes lineage commitment of lung epithelial progenitors by curbing Fgf10 and ß-catenin signaling. Our findings show that both inactivation of the Hippo pathway (nuclear Yap) or ablation of Yap result in increased ß-catenin and Fgf10 signaling, suggesting a cytoplasmic role for Yap in epithelial lineage commitment. We further demonstrate redundant and non-redundant functions for the two nuclear effectors of the Hippo pathway, Yap and Taz, during lung development.


Asunto(s)
Linaje de la Célula , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Pulmón/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Citoplasma/metabolismo , Femenino , Vía de Señalización Hippo , Pulmón/embriología , Masculino , Ratones , Modelos Biológicos , Organogénesis , Fenotipo , Fosfoproteínas/metabolismo , Alveolos Pulmonares/embriología , Transactivadores , Proteínas Señalizadoras YAP
15.
Nat Commun ; 9(1): 4600, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389913

RESUMEN

Impaired alveolar formation and maintenance are features of many pulmonary diseases that are associated with significant morbidity and mortality. In a forward genetic screen for modulators of mouse lung development, we identified the non-muscle myosin II heavy chain gene, Myh10. Myh10 mutant pups exhibit cyanosis and respiratory distress, and die shortly after birth from differentiation defects in alveolar epithelium and mesenchyme. From omics analyses and follow up studies, we find decreased Thrombospondin expression accompanied with increased matrix metalloproteinase activity in both mutant lungs and cultured mutant fibroblasts, as well as disrupted extracellular matrix (ECM) remodeling. Loss of Myh10 specifically in mesenchymal cells results in ECM deposition defects and alveolar simplification. Notably, MYH10 expression is downregulated in the lung of emphysema patients. Altogether, our findings reveal critical roles for Myh10 in alveologenesis at least in part via the regulation of ECM remodeling, which may contribute to the pathogenesis of emphysema.


Asunto(s)
Matriz Extracelular/metabolismo , Enfermedades Pulmonares/metabolismo , Cadenas Pesadas de Miosina/deficiencia , Miosina Tipo IIB no Muscular/deficiencia , Secuencia de Aminoácidos , Animales , Regulación hacia Abajo/genética , Enfisema/patología , Etilnitrosourea , Femenino , Enfermedades Pulmonares/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Mutagénesis/genética , Mutación Missense/genética , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo IIB no Muscular/química , Miosina Tipo IIB no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Organogénesis , Fenotipo , Alveolos Pulmonares/embriología , Alveolos Pulmonares/metabolismo , Regulación hacia Arriba/genética
16.
Blood ; 132(11): 1167-1179, 2018 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-29853539

RESUMEN

Platelets participate in not only thrombosis and hemostasis but also other pathophysiological processes, including tumor metastasis and inflammation. However, the putative role of platelets in the development of solid organs has not yet been described. Here, we report that platelets regulate lung development through the interaction between the platelet-activation receptor, C-type lectin-like receptor-2 (Clec-2; encoded by Clec1b), and its ligand, podoplanin, a membrane protein. Clec-2 deletion in mouse platelets led to lung malformation, which caused respiratory failure and neonatal lethality. In these embryos, α-smooth muscle actin-positive alveolar duct myofibroblasts (adMYFs) were almost absent in the primary alveolar septa, which resulted in loss of alveolar elastic fibers and lung malformation. Our data suggest that the lack of adMYFs is caused by abnormal differentiation of lung mesothelial cells (luMCs), the major progenitor of adMYFs. In the developing lung, podoplanin expression is detected in alveolar epithelial cells (AECs), luMCs, and lymphatic endothelial cells (LECs). LEC-specific podoplanin knockout mice showed neonatal lethality and Clec1b-/--like lung developmental abnormalities. Notably, these Clec1b-/--like lung abnormalities were also observed after thrombocytopenia or transforming growth factor-ß depletion in fetuses. We propose that the interaction between Clec-2 on platelets and podoplanin on LECs stimulates adMYF differentiation of luMCs through transforming growth factor-ß signaling, thus regulating normal lung development.


Asunto(s)
Plaquetas/metabolismo , Diferenciación Celular/fisiología , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Alveolos Pulmonares/embriología , Transducción de Señal/fisiología , Animales , Plaquetas/citología , Células Endoteliales , Células Epiteliales/citología , Células Epiteliales/metabolismo , Lectinas Tipo C/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Miofibroblastos/citología , Miofibroblastos/metabolismo , Alveolos Pulmonares/citología , Mucosa Respiratoria/citología , Mucosa Respiratoria/embriología
17.
Dev Biol ; 444 Suppl 1: S325-S336, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29792856

RESUMEN

Although the basic schema of the body plan is similar among different species of amniotes (mammals, birds, and reptiles), the lung is an exception. Here, anatomy and physiology are considerably different, particularly between mammals and birds. In mammals, inhaled and exhaled airs mix in the airways, whereas in birds the inspired air flows unidirectionally without mixing with the expired air. This bird-specific respiration system is enabled by the complex tubular structures called parabronchi where gas exchange takes place, and also by the bellow-like air sacs appended to the main part of the lung. That the lung is predominantly governed by the parasympathetic nervous system has been shown mostly by physiological studies in mammals. However, how the parasympathetic nervous system in the lung is established during late development has largely been unexplored both in mammals and birds. In this study, by combining immunocytochemistry, the tissue-clearing CUBIC method, and ink-injection to airways, we have visualized the 3-D distribution patterns of parasympathetic nerves and ganglia in the lung at late developmental stages of mice and chickens. These patterns were further compared between these species, and three prominent similarities emerged: (1) parasympathetic postganglionic fibers and ganglia are widely distributed in the lung covering the proximal and distal portions, (2) the gas exchange units, alveoli in mice and parabronchi in chickens, are devoid of parasympathetic nerves, (3) parasympathetic nerves are in close association with smooth muscle cells, particularly at the base of the gas exchange units. These observations suggest that despite gross differences in anatomy, the basic mechanisms underlying parasympathetic control of smooth muscles and gas exchange might be conserved between mammals and birds.


Asunto(s)
Pulmón/embriología , Pulmón/fisiología , Sistema Nervioso Parasimpático/fisiología , Animales , Embrión de Pollo , Pollos , Ganglios/embriología , Mamíferos/fisiología , Ratones , Ratones Endogámicos ICR , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Sistema Nervioso Parasimpático/embriología , Alveolos Pulmonares/embriología , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/fisiología
18.
Development ; 145(7)2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636361

RESUMEN

Platelet-derived growth factor A (PDGF-A) signaling through PDGF receptor α is essential for alveogenesis. Previous studies have shown that Pdgfa-/- mouse lungs have enlarged alveolar airspace with absence of secondary septation, both distinctive features of bronchopulmonary dysplasia. To study how PDGF-A signaling is involved in alveogenesis, we generated lung-specific Pdgfa knockout mice (Pdgfafl/-; Spc-cre) and characterized their phenotype postnatally. Histological differences between mutant mice and littermate controls were visible after the onset of alveogenesis and maintained until adulthood. Additionally, we generated Pdgfafl/-; Spc-cre; PdgfraGFP/+ mice in which Pdgfra+ cells exhibit nuclear GFP expression. In the absence of PDGF-A, the number of PdgfraGFP+ cells was significantly decreased. In addition, proliferation of PdgfraGFP+ cells was reduced. During alveogenesis, PdgfraGFP+ myofibroblasts failed to form the α-smooth muscle actin rings necessary for alveolar secondary septation. These results indicate that PDGF-A signaling is involved in myofibroblast proliferation and migration. In addition, we show an increase in both the number and proliferation of alveolar type II cells in Pdgfafl/-; Spc-cre lungs, suggesting that the increased alveolar airspace is not caused solely by deficient myofibroblast function.


Asunto(s)
Pulmón/metabolismo , Organogénesis/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Alveolos Pulmonares/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Alveolos Pulmonares/embriología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
19.
Am J Respir Crit Care Med ; 197(6): 776-787, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29268623

RESUMEN

RATIONALE: Pregnancies complicated by antenatal stress, including preeclampsia (PE) and chorioamnionitis (CA), increase the risk for bronchopulmonary dysplasia (BPD) in preterm infants, but biologic mechanisms linking prenatal factors with BPD are uncertain. Levels of sFlt-1 (soluble fms-like tyrosine kinase 1), an endogenous antagonist to VEGF (vascular endothelial growth factor), are increased in amniotic fluid and maternal blood in PE and associated with CA. OBJECTIVES: Because impaired VEGF signaling has been implicated in the pathogenesis of BPD, we hypothesized that fetal exposure to sFlt-1 decreases lung growth and causes abnormal lung structure and pulmonary hypertension during infancy. METHODS: To test this hypothesis, we studied the effects of anti-sFlt-1 monoclonal antibody (mAb) treatment on lung growth in two established antenatal models of BPD that mimic PE and CA induced by intraamniotic (i.a.) injections of sFlt-1 or endotoxin, respectively. In experimental PE, mAb was administered by three different approaches, including antenatal treatment by either i.a. instillation or maternal uterine artery infusion, or by postnatal intraperitoneal injections. RESULTS: With each strategy, mAb therapy improved infant lung structure as assessed by radial alveolar count, vessel density, right ventricular hypertrophy, and lung function. As found in the PE model, the adverse lung effects of i.a. endotoxin were also reduced by antenatal or postnatal mAb therapy. CONCLUSIONS: We conclude that treatment with anti-sFlt-1 mAb preserves lung structure and function and prevents right ventricular hypertrophy in two rat models of BPD of antenatal stress and speculate that early mAb therapy may provide a novel strategy for the prevention of BPD.


Asunto(s)
Displasia Broncopulmonar/fisiopatología , Endotelio Vascular/crecimiento & desarrollo , Pulmón/crecimiento & desarrollo , Alveolos Pulmonares/crecimiento & desarrollo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/embriología , Modelos Animales de Enfermedad , Endotelio Vascular/embriología , Femenino , Humanos , Pulmón/embriología , Embarazo , Alveolos Pulmonares/embriología , Ratas , Ratas Sprague-Dawley
20.
Cell Cycle ; 17(1): 80-91, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29143549

RESUMEN

Diabetes mellitus in pregnancy has been known to affect the embryonic development of various systems, including cardiovascular and nervous systems. However, whether this disease could have a negative impact on embryonic respiratory system remains controversial. In this study, we demonstrated that pregestational diabetes mellitus (PGDM)-induced defects in lung development in mice are mainly characterized by the changes in the morphological structure of the lung. Immunostaining and Western blotting showed that proliferation increased and apoptosis decreased in PGDM. Hyperglycaemia caused pulmonary tissue fibrationas manifested by an increase in Masson staining and decorin expression in PGDM lungs, and the immunofluorescent pro-SPC+ type II pulmonary epithelial cell number was decreased. The alteration of pulmonary epithelial cell differentiation might be due to hyperglycaemia-activated Wnt signalling and suppressed GATA6 expression in PGDM mouse lung tissues and MLE-12 cells. The treatment of MLE-12 cells with high glucose in the presence/absence of XAV939 or su5402 further proved that hyperglycaemia suppressed the expression of GATA6 and pro-SPC by activating Wnt signalling and induced the expression of decorin, α-SMA and TGF-ß by activating Fgf signalling. Therefore, in this study, we revealed that hyperglycemia induced dysfunctional pulmonary cell apoptosis and proliferation, as well as pulmonary myofibroblast hyperplasia, which contributed to the formation of aberrant structure of alveolar walls. Furthermore, the hyperglycaemia also inhibited the differentiation of pulmonary epithelial cells through the canonical Wnt and Fgf signalling, and the alteration of Fgf and Wnt signalling activated TGF-ß, which would promote the AECII EMT process.


Asunto(s)
Hiperglucemia/patología , Alveolos Pulmonares/embriología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Proliferación Celular , Células Epiteliales/patología , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción GATA6/metabolismo , Ratones , Modelos Biológicos , Alveolos Pulmonares/patología , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...