Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Chemistry ; 30(16): e202303733, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38055214

RESUMEN

Transition metal oxide cathodes (TMOCs) such as LiNi0.8Mn0.1Co0.1O2 and LiMn1.5Ni0.5O4 have been widely employed in Li-ion batteries (LIBs) owing to superior operating voltages, high reversible capacities and relatively low cost. Nevertheless, despite significant advancements in practical application, TMOC-based LIBs face great challenges such as transition metal dissolution and volume expansion during cycling, which jeopardizes the future advance of high-voltage TMOCs. As a critical component of cathode, polymeric binder acts as a crucial part in maintaining the mechanical and ion/electron conductive integrity between active particles, carbon additives, and the aluminum collector, hence minimizing cathode pulverization during battery cycling. Moreover, Polymeric binder with specialized functions is thought to offer a new solution to enhancing the electrochemical stability of the TMOCs. Therefore, this review aims at providing a comprehensive summary of the ideal requirements, design strategies and recent progress of polymeric binders for TMOCs. Future design perspectives and promising research technologies for advanced binders for high-voltage TMOCs are introduced.

2.
bioRxiv ; 2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37905051

RESUMEN

Alveolar epithelial regeneration is critical for normal lung function and becomes dysregulated in disease. While alveolar type 2 (AT2) and club cells are known distal lung epithelial progenitors, determining if alveolar epithelial type 1 (AT1) cells also contribute to alveolar regeneration has been hampered by lack of highly specific mouse models labeling AT1 cells. To address this, the Gramd2 CreERT2 transgenic strain was generated and crossed to Rosa mTmG mice. Extensive cellular characterization, including distal lung immunofluorescence and cytospin staining, confirmed that GRAMD2 + AT1 cells are highly enriched for green fluorescent protein (GFP). Interestingly, Gramd2 CreERT2 GFP + cells were able to form organoids in organoid co-culture with Mlg fibroblasts. Temporal scRNAseq revealed that Gramd2 + AT1 cells transition through numerous intermediate lung epithelial cell states including basal, secretory and AT2 cell in organoids while acquiring proliferative capacity. Our results indicate that Gramd2 + AT1 cells are highly plastic suggesting they may contribute to alveolar regeneration.

3.
Elife ; 112022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36214448

RESUMEN

Lung development is precisely controlled by underlying gene regulatory networks (GRN). Disruption of genes in the network can interrupt normal development and cause diseases such as bronchopulmonary dysplasia (BPD) - a chronic lung disease in preterm infants with morbid and sometimes lethal consequences characterized by lung immaturity and reduced alveolarization. Here, we generated a transgenic mouse exhibiting a moderate severity BPD phenotype by blocking IGF1 signaling in secondary crest myofibroblasts (SCMF) at the onset of alveologenesis. Using approaches mirroring the construction of the model GRN in sea urchin's development, we constructed the IGF1 signaling network underlying alveologenesis using this mouse model that phenocopies BPD. The constructed GRN, consisting of 43 genes, provides a bird's eye view of how the genes downstream of IGF1 are regulatorily connected. The GRN also reveals a mechanistic interpretation of how the effects of IGF1 signaling are transduced within SCMF from its specification genes to its effector genes and then from SCMF to its neighboring alveolar epithelial cells with WNT5A and FGF10 signaling as the bridge. Consistently, blocking WNT5A signaling in mice phenocopies BPD as inferred by the network. A comparative study on human samples suggests that a GRN of similar components and wiring underlies human BPD. Our network view of alveologenesis is transforming our perspective to understand and treat BPD. This new perspective calls for the construction of the full signaling GRN underlying alveologenesis, upon which targeted therapies for this neonatal chronic lung disease can be viably developed.


Asunto(s)
Displasia Broncopulmonar , Lactante , Humanos , Ratones , Recién Nacido , Animales , Displasia Broncopulmonar/genética , Redes Reguladoras de Genes , Recien Nacido Prematuro , Organogénesis , Modelos Animales de Enfermedad , Pulmón , Animales Recién Nacidos , Factor I del Crecimiento Similar a la Insulina/genética
4.
Cell Rep ; 39(1): 110608, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385750

RESUMEN

The lung alveolus is lined with alveolar type 1 (AT1) and type 2 (AT2) epithelial cells. During alveologenesis, increasing demand associated with expanding alveolar numbers is met by proliferating progenitor AT2s (pAT2). Little information exists regarding the identity of this population and their niche microenvironment. We show that during alveologenesis, Hedgehog-responsive PDGFRa(+) progenitors (also known as SCMFs) are a source of secreted trophic molecules that maintain a unique pAT2 population. SCMFs are in turn maintained by TGFß signaling. Compound inactivation of Alk5 TßR2 in SCMFs reduced their numbers and depleted the pAT2 pool without impacting differentiation of daughter cells. In lungs of preterm infants who died with bronchopulmonary dysplasia, PDGFRa is reduced and the number of proliferative AT2s is diminished, indicating that an evolutionarily conserved mechanism governs pAT2 behavior during alveologenesis. SCMFs are a transient cell population, active only during alveologenesis, making them a unique stage-specific niche mesodermal cell type in mammalian organs.


Asunto(s)
Erizos , Recien Nacido Prematuro , Animales , Diferenciación Celular/fisiología , Células Epiteliales , Fibroblastos , Humanos , Recién Nacido , Pulmón , Organogénesis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células Madre/metabolismo
5.
Stem Cells ; 40(7): 691-703, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35429397

RESUMEN

Lung maturation is not limited to proper structural development but also includes differentiation and functionality of various highly specialized alveolar cell types. Alveolar type 1 (AT1s) cells occupy nearly 95% of the alveolar surface and are critical for establishing efficient gas exchange in the mature lung. AT1 cells arise from progenitors specified during the embryonic stage as well as alveolar epithelial progenitors expressing surfactant protein C (Sftpcpos cells) during postnatal and adult stages. Previously, we found that Wnt5a, a non-canonical Wnt ligand, is required for differentiation of AT1 cells during the saccular phase of lung development. To further investigate the role of Wnt5a in AT1 cell differentiation, we generated and characterized a conditional Wnt5a gain-of-function mouse model. Neonatal Wnt5a gain-of-function disrupted alveologenesis through inhibition of cell proliferation. In this setting Wnt5a downregulated ß-catenin-dependent canonical Wnt signaling, repressed AT2 (anti-AT2) and promoted AT1 (pro-AT1) lineage-specific gene expression. In addition, we identified 2 subpopulations of Sftpchigh and Sftpclow alveolar epithelial cells. In Sftpclow cells, Wnt5a exhibits pro-AT1 and anti-AT2 effects, concurrent with inhibition of canonical Wnt signaling. Interestingly, in the Sftpchigh subpopulation, although increasing AT1 lineage-specific gene expression, Wnt5a gain-of-function did not change AT2 gene expression, nor inhibit canonical Wnt signaling. Using primary epithelial cells isolated from human fetal lungs, we demonstrate that this property of Wnt5a is evolutionarily conserved. Wnt5a therefore serves as a selective regulator that ensures proper AT1/AT2 balance in the developing lung.


Asunto(s)
Células Epiteliales Alveolares , Vía de Señalización Wnt , Células Epiteliales Alveolares/metabolismo , Animales , Diferenciación Celular/genética , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Recién Nacido , Ratones , Vía de Señalización Wnt/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
6.
Cells ; 11(7)2022 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-35406686

RESUMEN

NKX2.1 is a master regulator of lung morphogenesis and cell specification; however, interactions of NKX2.1 with various transcription factors to regulate cell-specific gene expression and cell fate in the distal lung remain incompletely understood. FOXO1 is a key regulator of stem/progenitor cell maintenance/differentiation in several tissues but its role in the regulation of lung alveolar epithelial progenitor homeostasis has not been evaluated. We identified a novel role for FOXO1 in alveolar epithelial cell (AEC) differentiation that results in the removal of NKX2.1 from surfactant gene promoters and the subsequent loss of surfactant expression in alveolar epithelial type I-like (AT1-like) cells. We found that the FOXO1 forkhead domain potentiates a loss of surfactant gene expression through an interaction with the NKX2.1 homeodomain, disrupting NKX2.1 binding to the SFTPC promoter. In addition, blocking PI-3K/AKT signaling reduces phosphorylated FOXO-1 (p-FOXO1), allowing accumulated nuclear FOXO1 to interact with NKX2.1 in differentiating AEC. Inhibiting AEC differentiation in vitro with keratinocyte growth factor (KGF) maintained an AT2 cell phenotype through increased PI3K/AKT-mediated FOXO1 phosphorylation, resulting in higher levels of surfactant expression. Together these results indicate that FOXO1 plays a central role in AEC differentiation by directly binding NKX2.1 and suggests an essential role for FOXO1 in mediating AEC homeostasis.


Asunto(s)
Células Epiteliales Alveolares , Surfactantes Pulmonares , Células Epiteliales Alveolares/metabolismo , Células Epiteliales/metabolismo , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Surfactantes Pulmonares/metabolismo , Tensoactivos/metabolismo
7.
Org Lett ; 22(17): 6842-6846, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32810404

RESUMEN

The classical Sonogashira reaction of aryl electrophiles in the presence of Pd catalysts has been well established as a potent method for arylalkyne synthesis. However, the site-selective C(sp2)-C(sp) cross-coupling strategy using a non-noble-metal catalyst is rare. An efficient alternative approach for the synthesis of arylalkynes via a Cu-catalyzed Sonogashira-type reaction promoted by visible light is described. This method enables site-selective alkynylation from aryl sulfonium salts derived from diverse arenes to a set of arylalkynes with high selectivity and high functional-group compatibility. Moreover, rapid alkynylation of drug molecules is demonstrated.

8.
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
9.
Am J Respir Crit Care Med ; 201(2): 198-211, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31738079

RESUMEN

Rationale: Alveolar epithelial cell (AEC) injury and dysregulated repair are implicated in the pathogenesis of pulmonary fibrosis. Endoplasmic reticulum (ER) stress in AEC has been observed in idiopathic pulmonary fibrosis (IPF), a disease of aging.Objectives: To investigate a causal role for ER stress in the pathogenesis of pulmonary fibrosis (PF) and therapeutic potential of ER stress inhibition in PF.Methods: The role of ER stress in AEC dysfunction and fibrosis was studied in mice with tamoxifen (Tmx)-inducible deletion of ER chaperone Grp78, a key regulator of ER homeostasis, in alveolar type II (AT2) cells, progenitors of distal lung epithelium, and in IPF lung slice cultures.Measurements and Main Results:Grp78 deletion caused weight loss, mortality, lung inflammation, and spatially heterogeneous fibrosis characterized by fibroblastic foci, hyperplastic AT2 cells, and increased susceptibility of old and male mice, all features of IPF. Fibrosis was more persistent in more severely injured Grp78 knockout (KO) mice. Grp78 KO AT2 cells showed evidence of ER stress, apoptosis, senescence, impaired progenitor capacity, and activation of TGF-ß (transforming growth factor-ß)/SMAD signaling. Glucose-regulated protein 78 is reduced in AT2 cells from old mice and patients with IPF, and ER stress inhibitor tauroursodeoxycholic acid ameliorates ER stress and fibrosis in Grp78 KO mouse and IPF lung slice cultures.Conclusions: These results support a causal role for ER stress and resulting epithelial dysfunction in PF and suggest ER stress as a potential mechanism linking aging to IPF. Modulation of ER stress and chaperone function may offer a promising therapeutic approach for pulmonary fibrosis.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Estrés del Retículo Endoplásmico/genética , Proteínas de Choque Térmico/genética , Fibrosis Pulmonar/genética , Células Madre/metabolismo , Factores de Edad , Células Epiteliales Alveolares/patología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/genética , Senescencia Celular/genética , Dasatinib/farmacología , Chaperón BiP del Retículo Endoplásmico , Técnicas de Inactivación de Genes , Proteínas de Choque Térmico/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/efectos de los fármacos , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Quercetina/farmacología , Quinolinas/farmacología , Proteínas Smad/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Factor de Transcripción CHOP/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
10.
Stem Cell Res Ther ; 10(1): 366, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31791401

RESUMEN

BACKGROUND: The airways of mammalian lung are lined with highly specialized cell types that are the target of airborne toxicants and injury. Several epithelial cell types and bone marrow-derived mesenchymal stem cells have been identified to serve as stem cells during injury repair. However, the contributions of endogenous mesenchymal cells to recruitment, expansion or differentiation of stem cells, and repair and reestablishment of the normal composition of airway epithelium following injury have not been addressed. METHODS: The role of mouse pulmonary mesenchymal cells was investigated by lineage tracing using Dermo1-Cre; ROSAmTmG mice. In experimental models of lung injury by lipopolysaccharide and naphthalene, GFP-labeled Dermo1+ mesenchymal cells were traced during injury repair. In vitro lung explant culture treated with or without lipopolysaccharide was also used to verify in vivo data. RESULTS: During injury repair, a subgroup of GFP-labeled Dermo1+ mesenchymal cells were found to contribute to normal repair of the airway epithelium and differentiated into Club cells, ciliated cells, and goblet cells. In Club cell-specific naphthalene injury model, the process of Dermo1+ stem cell regenerating epithelial cells was dissected. The Dermo1+ stem cells was migrated into the airway epithelium layer sooner after injury, and sequentially differentiated transitionally to epithelial stem cells, such as neuroendocrine cells, and finally to newly differentiated Club cells, ciliated cells, and goblet cells in injury repair. CONCLUSION: In this study, a population of Dermo1+ mesenchymal stem cell was identified to serve as stem cells in airway epithelial cell regeneration during injury repair. The Dermo1+ mesenchymal stem cell differentiated into epithelial stem cells before reestablishing various epithelial cells. These findings have implications for understanding the regulation of lung repair and the potential for usage of mesenchymal stem cells in therapeutic strategies for lung diseases.


Asunto(s)
Epitelio/fisiología , Lesión Pulmonar/terapia , Trasplante de Células Madre Mesenquimatosas , Regeneración , Enfermedad Aguda , Animales , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Lipopolisacáridos/toxicidad , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Lesión Pulmonar/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Transgénicos , Naftalenos/toxicidad , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
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.
Sci Rep ; 7(1): 3473, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615712

RESUMEN

Epigenetic regulation of differentiation-related genes is poorly understood. We previously reported that transcription factors GATA6 and Sp1 interact with and activate the rat proximal 358-bp promoter/enhancer (p358P/E) of lung alveolar epithelial type I (AT1) cell-specific gene aquaporin-5 (Aqp5). In this study, we found that histone deacetylase (HDAC) inhibitor suberoylanilide hydroxamic acid (SAHA) increased AQP5 expression and Sp1-mediated transcription of p358P/E. HDAC3 overexpression inhibited Sp1-mediated Aqp5 activation, while HDAC3 knockdown augmented AQP5 protein expression. Knockdown of GATA6 or transcriptional co-activator/histone acetyltransferase p300 decreased AQP5 expression, while p300 overexpression enhanced p358P/E activation by GATA6 and Sp1. GATA6 overexpression, SAHA treatment or HDAC3 knockdown increased histone H3 (H3) but not histone H4 (H4) acetylation within the homologous p358P/E region of mouse Aqp5. HDAC3 binds to Sp1 and HDAC3 knockdown increased interaction of GATA6/Sp1, GATA6/p300 and Sp1/p300. These results indicate that GATA6 and HDAC3 control Aqp5 transcription via modulation of H3 acetylation/deacetylation, respectively, through competition for binding to Sp1, and suggest that p300 modulates acetylation and/or interacts with GATA6/Sp1 to regulate Aqp5 transcription. Cooperative interactions among transcription factors and histone modifications regulate Aqp5 expression during alveolar epithelial cell transdifferentiation, suggesting that HDAC inhibitors may enhance repair by promoting acquisition of AT1 cell phenotype.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Acuaporina 5/genética , Factor de Transcripción GATA6/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Factor de Transcripción Sp1/metabolismo , Acetilación , Animales , Línea Celular , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Ratones , Modelos Biológicos , Factores de Transcripción p300-CBP/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 312(3): L371-L391, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28062486

RESUMEN

Various pathophysiological conditions such as surfactant dysfunction, mechanical ventilation, inflammation, pathogen products, environmental exposures, and gastric acid aspiration stress lung cells, and the compromise of plasma membranes occurs as a result. The mechanisms necessary for cells to repair plasma membrane defects have been extensively investigated in the last two decades, and some of these key repair mechanisms are also shown to occur following lung cell injury. Because it was theorized that lung wounding and repair are involved in the pathogenesis of acute respiratory distress syndrome (ARDS) and idiopathic pulmonary fibrosis (IPF), in this review, we summarized the experimental evidence of lung cell injury in these two devastating syndromes and discuss relevant genetic, physical, and biological injury mechanisms, as well as mechanisms used by lung cells for cell survival and membrane repair. Finally, we discuss relevant signaling pathways that may be activated by chronic or repeated lung cell injury as an extension of our cell injury and repair focus in this review. We hope that a holistic view of injurious stimuli relevant for ARDS and IPF could lead to updated experimental models. In addition, parallel discussion of membrane repair mechanisms in lung cells and injury-activated signaling pathways would encourage research to bridge gaps in current knowledge. Indeed, deep understanding of lung cell wounding and repair, and discovery of relevant repair moieties for lung cells, should inspire the development of new therapies that are likely preventive and broadly effective for targeting injurious pulmonary diseases.


Asunto(s)
Membrana Celular/patología , Enfermedades Pulmonares/patología , Cicatrización de Heridas , Animales , Humanos , Lesión Pulmonar/patología , Modelos Biológicos , Transducción de Señal
14.
BMC Biol ; 14: 19, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26984772

RESUMEN

BACKGROUND: Epithelial-mesenchymal cross talk is centerpiece in the development of many branched organs, including the lungs. The embryonic lung mesoderm provides instructional information not only for lung architectural development, but also for patterning, commitment and differentiation of its many highly specialized cell types. The mesoderm also serves as a reservoir of progenitors for generation of differentiated mesenchymal cell types that include αSMA-expressing fibroblasts, lipofibroblasts, endothelial cells and others. Transforming Growth Factor ß (TGFß) is a key signaling pathway in epithelial-mesenchymal cross talk. Using a cre-loxP approach we have elucidated the role of the TGFß type I receptor tyrosine kinase, ALK5, in epithelial-mesenchymal cross talk during lung morphogenesis. RESULTS: Targeted early inactivation of Alk5 in mesodermal progenitors caused abnormal development and maturation of the lung that included reduced physical size of the sub-mesothelial mesoderm, an established source of specific mesodermal progenitors. Abrogation of mesodermal ALK5-mediated signaling also inhibited differentiation of cell populations in the epithelial and endothelial lineages. Importantly, Alk5 mutant lungs contained a reduced number of αSMA(pos) cells and correspondingly increased lipofibroblasts. Elucidation of the underlying mechanisms revealed that through direct and indirect modulation of target signaling pathways and transcription factors, including PDGFRα, PPARγ, PRRX1, and ZFP423, ALK5-mediated TGFß controls a process that regulates the commitment and differentiation of αSMA(pos) versus lipofibroblast cell populations during lung development. CONCLUSION: ALK5-mediated TGFß signaling controls an early pathway that regulates the commitment and differentiation of αSMA(pos) versus LIF cell lineages during lung development.


Asunto(s)
Pulmón/citología , Pulmón/embriología , Mesodermo/citología , Mesodermo/embriología , Miofibroblastos/citología , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Células Madre/citología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Pulmón/anomalías , Pulmón/metabolismo , Mesodermo/anomalías , Mesodermo/metabolismo , Ratones Endogámicos C57BL , Músculo Liso/anomalías , Músculo Liso/citología , Músculo Liso/embriología , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Células Madre/metabolismo , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo
15.
J Biol Chem ; 291(12): 6569-82, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26833564

RESUMEN

Maintenance of stem/progenitor cell-progeny relationships is required for tissue homeostasis during normal turnover and repair. Wnt signaling is implicated in both maintenance and differentiation of adult stem/progenitor cells, yet how this pathway serves these dichotomous roles remains enigmatic. We previously proposed a model suggesting that specific interaction of ß-catenin with either of the homologous Kat3 co-activators, p300 or CREB-binding protein, differentially regulates maintenance versus differentiation of embryonic stem cells. Limited knowledge of endogenous mechanisms driving differential ß-catenin/co-activator interactions and their role in adult somatic stem/progenitor cell maintenance versus differentiation led us to explore this process in defined models of adult progenitor cell differentiation. We focused primarily on alveolar epithelial type II (AT2) cells, progenitors of distal lung epithelium, and identified a novel axis whereby WNT5a/protein kinase C (PKC) signaling regulates specific ß-catenin/co-activator interactions to promote adult progenitor cell differentiation. p300/ß-catenin but not CBP/ß-catenin interaction increases as AT2 cells differentiate to a type I (AT1) cell-like phenotype. Additionally, p300 transcriptionally activates AT1 cell-specific gene Aqp-5. IQ-1, a specific inhibitor of p300/ß-catenin interaction, prevents differentiation of not only primary AT2 cells, but also tracheal epithelial cells, and C2C12 myoblasts. p300 phosphorylation at Ser-89 enhances p300/ß-catenin interaction, concurrent with alveolar epithelial cell differentiation. WNT5a, a traditionally non-canonical WNT ligand regulates Ser-89 phosphorylation and p300/ß-catenin interactions in a PKC-dependent manner, likely involving PKCζ. These studies identify a novel intersection of canonical and non-canonical Wnt signaling in adult progenitor cell differentiation that has important implications for targeting ß-catenin to modulate adult progenitor cell behavior in disease.


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular , Proteína p300 Asociada a E1A/fisiología , Proteína Quinasa C/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/fisiología , Células Epiteliales Alveolares/fisiología , Animales , Acuaporina 5/genética , Acuaporina 5/metabolismo , Línea Celular , Impedancia Eléctrica , Expresión Génica , Ratones , Ratones Noqueados , Fosforilación , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Ratas , Vía de Señalización Wnt , Proteína Wnt-5a
16.
Am J Respir Cell Mol Biol ; 55(1): 135-49, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26816051

RESUMEN

Bronchopulmonary dysplasia (BPD), a chronic lung disease of prematurity, has been linked to endoplasmic reticulum (ER) stress. To investigate a causal role for ER stress in BPD pathogenesis, we generated conditional knockout (KO) mice (cGrp78(f/f)) with lung epithelial cell-specific KO of Grp78, a gene encoding the ER chaperone 78-kD glucose-regulated protein (GRP78), a master regulator of ER homeostasis and the unfolded protein response (UPR). Lung epithelial-specific Grp78 KO disrupted lung morphogenesis, causing developmental arrest, increased alveolar epithelial type II cell apoptosis, and decreased surfactant protein and type I cell marker expression in perinatal lungs. cGrp78(f/f) pups died immediately after birth, likely owing to respiratory distress. Importantly, Grp78 KO triggered UPR activation with marked induction of the proapoptotic transcription factor CCAAT/enhancer-binding proteins (C/EBP) homologous protein (CHOP). Increased expression of genes involved in oxidative stress and cell death and decreased expression of genes encoding antioxidant enzymes suggest a role for oxidative stress in alveolar epithelial cell (AEC) apoptosis. Increased Smad3 phosphorylation and expression of transforming growth factor-ß/Smad3 targets Cdkn1a (encoding p21) and Gadd45a suggest that interactions among the apoptotic arm of the UPR, oxidative stress, and transforming growth factor-ß/Smad signaling pathways contribute to Grp78 KO-induced AEC apoptosis and developmental arrest. Chemical chaperone Tauroursodeoxycholic acid reduced UPR activation and apoptosis in cGrp78(f/f) lungs cultured ex vivo, confirming a role for ER stress in observed AEC abnormalities. These results demonstrate a key role for GRP78 in AEC survival and gene expression during lung development through modulation of ER stress, and suggest the UPR as a potential therapeutic target in BPD.


Asunto(s)
Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Homeostasis , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/ultraestructura , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Homeostasis/efectos de los fármacos , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
17.
Dev Biol ; 408(1): 56-65, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26460096

RESUMEN

Pten is a multifunctional tumor suppressor. Deletions and mutations in the Pten gene have been associated with multiple forms of human cancers. Pten is a central regulator of several signaling pathways that influences multiple cellular functions. One such function is in cell motility and migration, although the precise mechanism remains unknown. In this study, we deleted Pten in the embryonic lung epithelium using Gata5-cre mice. Absence of Pten blocked branching morphogenesis and ERK and AKT phosphorylation at E12.5. In an explant model, Pten(Δ/Δ) mesenchyme-free embryonic lung endoderm failed to branch. Inhibition of budding in Pten(Δ/Δ) explants was associated with major changes in cell migration, while cell proliferation was not affected. We further examined the role of ERK and AKT in branching morphogenesis by conditional, endodermal-specific mutants which blocked ERK or AKT phosphorylation. MEK(DM/+); Gata5-cre (blocking of ERK phosphorylation) lung showed more severe phenotype in branching morphogenesis. The inhibition of budding was also associated with disruption of cell migration. Thus, the mechanisms by which Pten is required for early endodermal morphogenesis may involve ERK, but not AKT, mediated cell migration.


Asunto(s)
Endodermo/embriología , Endodermo/enzimología , Pulmón/embriología , Sistema de Señalización de MAP Quinasas , Morfogénesis , Fosfohidrolasa PTEN/metabolismo , Animales , Movimiento Celular , Epitelio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor de Transcripción GATA5/metabolismo , Eliminación de Gen , Integrasas/metabolismo , Ratones , Modelos Biológicos , Especificidad de Órganos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
18.
J Biochem ; 158(5): 355-65, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26261051

RESUMEN

The role of WNT signalling in metazoan organogenesis has been a topic of widespread interest. In the lung, while the role of canonical WNT signalling has been examined in some detail by multiple studies, the non-canonical WNT signalling has received limited attention. Reliable evidence shows that this important signalling mechanism constitutes a major regulatory pathway in lung development. In addition, accumulating evidence has also shown that the non-canonical WNT pathway is critical for maintaining lung homeostasis and that aberrant activation of this pathway may underlie several debilitating lung diseases. Functional analyses have further revealed that the non-canonical WNT pathway regulates multiple cellular activities in the lung that are dependent on the specific cellular context. In most cell types, non-canonical WNT signalling regulates canonical WNT activity, which is also critical for many aspects of lung biology. This review will summarize what is currently known about the role of non-canonical WNT signalling in lung development, homeostasis and pathogenesis of disease.


Asunto(s)
Pulmón/metabolismo , Modelos Biológicos , Organogénesis , Mucosa Respiratoria/metabolismo , Vía de Señalización Wnt , Animales , Madurez de los Órganos Fetales , Humanos , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Pulmón/patología , Enfermedades Pulmonares/inmunología , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Mucosa Respiratoria/embriología , Mucosa Respiratoria/crecimiento & desarrollo , Mucosa Respiratoria/patología
19.
Stem Cells ; 33(3): 999-1012, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25448080

RESUMEN

Development of the mammalian lung is predicated on cross-communications between two highly interactive tissues, the endodermally derived epithelium and the mesodermally derived pulmonary mesenchyme. While much attention has been paid for the lung epithelium, the pulmonary mesenchyme, partly due to lack of specific tractable markers remains under-investigated. The lung mesenchyme is derived from the lateral plate mesoderm and is the principal recipient of Hedgehog (Hh) signaling, a morphogenetic network that regulates multiple aspects of embryonic development. Using the Hh-responsive Gli1-cre(ERT2) mouse line, we identified the mesodermal targets of Hh signaling at various time points during embryonic and postnatal lung development. Cell lineage analysis showed these cells serve as progenitors to contribute to multiple lineages of mesodermally derived differentiated cell types that include parenchymal or interstitial myofibroblasts, peribronchial and perivascular smooth muscle as well as rare populations of cells within the mesothelium. Most importantly, Gli1-cre(ERT2) identified the progenitors of secondary crest myofibroblasts, a hitherto intractable cell type that plays a key role in alveolar formation, a vital process about which little is currently known. Transcriptome analysis of Hh-targeted progenitor cells transitioning from the pseudoglandular to the saccular phase of lung development revealed important modulations of key signaling pathways. Among these, there was significant downregulation of canonical WNT signaling. Ectopic stabilization of ß-catenin via inactivation of Apc by Gli1-cre(ERT2) expanded the Hh-targeted progenitor pools, which caused the formation of fibroblastic masses within the lung parenchyma. The Gli1-cre(ERT2) mouse line represents a novel tool in the analysis of mesenchymal cell biology and alveolar formation during lung development.


Asunto(s)
Pulmón/embriología , Mesodermo/citología , Miofibroblastos/citología , Células Madre/citología , Animales , Diferenciación Celular/fisiología , Femenino , Perfilación de la Expresión Génica , Pulmón/citología , Mesodermo/metabolismo , Ratones , Miofibroblastos/metabolismo , Embarazo , Transducción de Señal , Células Madre/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 306(4): L316-25, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24375794

RESUMEN

Many signaling pathways are mediated by Shc adapter proteins that, in turn, are expressed as three isoforms with distinct functions. The p66(Shc) isoform antagonizes proliferation, regulates oxidative stress, and mediates apoptosis. It is highly expressed in the canalicular but not the later stages of mouse lung development, and its expression persists in bronchopulmonary dysplasia, a chronic disease associated with premature birth. These observations suggest that p66(Shc) has a developmental function. However, constitutive p66(Shc) deletion yields no morphological phenotype, and the structure of the Shc gene precludes its inducible deletion. To elucidate its function in lung development, we transfected p66(Shc) or nonsilencing small-interfering RNA (siRNA) into the epithelia of embryonic day 11 mouse lungs that were then cultured for 3 days and analyzed morphometrically. To assess cellular proliferation and epithelial differentiation, lung explants were immunostained and immunoblotted for p66(Shc), proliferating cell nuclear antigen (PCNA), the proximal airway differentiation antigens Clara cell 10-kDa protein (CC10) and thyroid transcription factor (TTF)-1, and the alveolar surfactant proteins (SP)-A, -B, and -C. Explants transfected with nonsilencing siRNA demonstrated specific epithelial uptake and normal morphological development relative to uninjected controls. In contrast, transfection with p66(Shc) siRNA significantly increased lumenal cross-sectional areas, decreased branching, and increased epithelial proliferation (P < 0.05 for all). Relative to controls, the expression of SP-B, SP-C, CC10, and TTF-1 was decreased by p66(Shc) knockdown. SP-A was not expressed in either control or treated lungs. These data suggest that p66(Shc) attenuates epithelial proliferation while promoting both distal and proximal epithelial maturation.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Pulmón/embriología , Morfogénesis , Proteínas Adaptadoras de la Señalización Shc/fisiología , Células 3T3 , Animales , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Pulmón/citología , Pulmón/metabolismo , Ratones , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína B Asociada a Surfactante Pulmonar/metabolismo , ARN Interferente Pequeño/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Técnicas de Cultivo de Tejidos , Factores de Transcripción , Uteroglobina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...