Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.612
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(10): 2428-2445.e20, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38579712

RESUMEN

Alveolar type 2 (AT2) cells are stem cells of the alveolar epithelia. Previous genetic lineage tracing studies reported multiple cellular origins for AT2 cells after injury. However, conventional lineage tracing based on Cre-loxP has the limitation of non-specific labeling. Here, we introduced a dual recombinase-mediated intersectional genetic lineage tracing approach, enabling precise investigation of AT2 cellular origins during lung homeostasis, injury, and repair. We found AT1 cells, being terminally differentiated, did not contribute to AT2 cells after lung injury and repair. Distinctive yet simultaneous labeling of club cells, bronchioalveolar stem cells (BASCs), and existing AT2 cells revealed the exact contribution of each to AT2 cells post-injury. Mechanistically, Notch signaling inhibition promotes BASCs but impairs club cells' ability to generate AT2 cells during lung repair. This intersectional genetic lineage tracing strategy with enhanced precision allowed us to elucidate the physiological role of various epithelial cell types in alveolar regeneration following injury.


Asunto(s)
Células Epiteliales Alveolares , Pulmón , Células Madre , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Diferenciación Celular , Linaje de la Célula , Pulmón/citología , Pulmón/metabolismo , Pulmón/fisiología , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Receptores Notch/metabolismo , Regeneración , Transducción de Señal , Células Madre/metabolismo , Células Madre/citología
2.
Cell ; 180(1): 107-121.e17, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31866069

RESUMEN

Fibrosis can develop in most organs and causes organ failure. The most common type of lung fibrosis is known as idiopathic pulmonary fibrosis, in which fibrosis starts at the lung periphery and then progresses toward the lung center, eventually causing respiratory failure. Little is known about the mechanisms underlying the pathogenesis and periphery-to-center progression of the disease. Here we discovered that loss of Cdc42 function in alveolar stem cells (AT2 cells) causes periphery-to-center progressive lung fibrosis. We further show that Cdc42-null AT2 cells in both post-pneumonectomy and untreated aged mice cannot regenerate new alveoli, resulting in sustained exposure of AT2 cells to elevated mechanical tension. We demonstrate that elevated mechanical tension activates a TGF-ß signaling loop in AT2 cells, which drives the periphery-to-center progression of lung fibrosis. Our study establishes a direct mechanistic link between impaired alveolar regeneration, mechanical tension, and progressive lung fibrosis.


Asunto(s)
Células Madre Adultas/metabolismo , Fibrosis Pulmonar Idiopática/etiología , Alveolos Pulmonares/metabolismo , Células Madre Adultas/patología , Anciano , Células Epiteliales Alveolares/patología , Animales , Fenómenos Biomecánicos/fisiología , Femenino , Fibrosis/patología , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Masculino , Ratones , Persona de Mediana Edad , Alveolos Pulmonares/patología , Regeneración , Transducción de Señal , Células Madre/patología , Estrés Mecánico , Estrés Fisiológico/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
3.
Nature ; 631(8021): 627-634, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38987592

RESUMEN

Fibroblasts are present throughout the body and function to maintain tissue homeostasis. Recent studies have identified diverse fibroblast subsets in healthy and injured tissues1,2, but the origins and functional roles of injury-induced fibroblast lineages remain unclear. Here we show that lung-specialized alveolar fibroblasts take on multiple molecular states with distinct roles in facilitating responses to fibrotic lung injury. We generate a genetic tool that uniquely targets alveolar fibroblasts to demonstrate their role in providing niches for alveolar stem cells in homeostasis and show that loss of this niche leads to exaggerated responses to acute lung injury. Lineage tracing identifies alveolar fibroblasts as the dominant origin for multiple emergent fibroblast subsets sequentially driven by inflammatory and pro-fibrotic signals after injury. We identify similar, but not completely identical, fibroblast lineages in human pulmonary fibrosis. TGFß negatively regulates an inflammatory fibroblast subset that emerges early after injury and stimulates the differentiation into fibrotic fibroblasts to elicit intra-alveolar fibrosis. Blocking the induction of fibrotic fibroblasts in the alveolar fibroblast lineage abrogates fibrosis but exacerbates lung inflammation. These results demonstrate the multifaceted roles of the alveolar fibroblast lineage in maintaining normal alveolar homeostasis and orchestrating sequential responses to lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Linaje de la Célula , Fibroblastos , Neumonía , Alveolos Pulmonares , Fibrosis Pulmonar , Animales , Femenino , Humanos , Masculino , Ratones , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/metabolismo , Diferenciación Celular , Fibroblastos/patología , Fibroblastos/metabolismo , Homeostasis , Neumonía/patología , Neumonía/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Células Madre/citología , Células Madre/patología , Factor de Crecimiento Transformador beta/metabolismo
4.
Cell ; 156(3): 440-55, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485453

RESUMEN

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Asunto(s)
Bronquiolos/citología , Diferenciación Celular , Células Endoteliales/metabolismo , Alveolos Pulmonares/citología , Transducción de Señal , Células Madre/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Bronquiolos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ratones , Factores de Transcripción NFATC/metabolismo , Alveolos Pulmonares/metabolismo , Células Madre/citología , Trombospondina 1/genética , Trombospondina 1/metabolismo
5.
Nature ; 621(7980): 813-820, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37587341

RESUMEN

Disruption of the lung endothelial-epithelial cell barrier following respiratory virus infection causes cell and fluid accumulation in the air spaces and compromises vital gas exchange function1. Endothelial dysfunction can exacerbate tissue damage2,3, yet it is unclear whether the lung endothelium promotes host resistance against viral pathogens. Here we show that the environmental sensor aryl hydrocarbon receptor (AHR) is highly active in lung endothelial cells and protects against influenza-induced lung vascular leakage. Loss of AHR in endothelia exacerbates lung damage and promotes the infiltration of red blood cells and leukocytes into alveolar air spaces. Moreover, barrier protection is compromised and host susceptibility to secondary bacterial infections is increased when endothelial AHR is missing. AHR engages tissue-protective transcriptional networks in endothelia, including the vasoactive apelin-APJ peptide system4, to prevent a dysplastic and apoptotic response in airway epithelial cells. Finally, we show that protective AHR signalling in lung endothelial cells is dampened by the infection itself. Maintenance of protective AHR function requires a diet enriched in naturally occurring AHR ligands, which activate disease tolerance pathways in lung endothelia to prevent tissue damage. Our findings demonstrate the importance of endothelial function in lung barrier immunity. We identify a gut-lung axis that affects lung damage following encounters with viral pathogens, linking dietary composition and intake to host fitness and inter-individual variations in disease outcome.


Asunto(s)
Células Endoteliales , Pulmón , Infecciones por Orthomyxoviridae , Receptores de Hidrocarburo de Aril , Animales , Humanos , Ratones , Apelina/metabolismo , Dieta , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Células Epiteliales/metabolismo , Eritrocitos/metabolismo , Gripe Humana/inmunología , Gripe Humana/metabolismo , Intestinos/metabolismo , Leucocitos/metabolismo , Ligandos , Pulmón/inmunología , Pulmón/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
6.
Development ; 151(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38602485

RESUMEN

Alveologenesis, the final stage in lung development, substantially remodels the distal lung, expanding the alveolar surface area for efficient gas exchange. Secondary crest myofibroblasts (SCMF) exist transiently in the neonatal distal lung and are crucial for alveologenesis. However, the pathways that regulate SCMF function, proliferation and temporal identity remain poorly understood. To address this, we purified SCMFs from reporter mice, performed bulk RNA-seq and found dynamic changes in Hippo-signaling components during alveologenesis. We deleted the Hippo effectors Yap/Taz from Acta2-expressing cells at the onset of alveologenesis, causing a significant arrest in alveolar development. Using single cell RNA-seq, we identified a distinct cluster of cells in mutant lungs with altered expression of marker genes associated with proximal mesenchymal cell types, airway smooth muscle and alveolar duct myofibroblasts. In vitro studies confirmed that Yap/Taz regulates myofibroblast-associated gene signature and contractility. Together, our findings show that Yap/Taz is essential for maintaining functional myofibroblast identity during postnatal alveologenesis.


Asunto(s)
Diferenciación Celular , Vía de Señalización Hippo , Morfogénesis , Miofibroblastos , Proteínas Serina-Treonina Quinasas , Alveolos Pulmonares , Transducción de Señal , Proteínas Señalizadoras YAP , Animales , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/citología , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Morfogénesis/genética , Mesodermo/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Pulmón/metabolismo , Organogénesis/genética , Regulación del Desarrollo de la Expresión Génica
7.
Nature ; 586(7831): 785-789, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33057196

RESUMEN

In the mammalian lung, an apparently homogenous mesh of capillary vessels surrounds each alveolus, forming the vast respiratory surface across which oxygen transfers to the blood1. Here we use single-cell analysis to elucidate the cell types, development, renewal and evolution of the alveolar capillary endothelium. We show that alveolar capillaries are mosaics; similar to the epithelium that lines the alveolus, the alveolar endothelium is made up of two intermingled cell types, with complex 'Swiss-cheese'-like morphologies and distinct functions. The first cell type, which we term the 'aerocyte', is specialized for gas exchange and the trafficking of leukocytes, and is unique to the lung. The other cell type, termed gCap ('general' capillary), is specialized to regulate vasomotor tone, and functions as a stem/progenitor cell in capillary homeostasis and repair. The two cell types develop from bipotent progenitors, mature gradually and are affected differently in disease and during ageing. This cell-type specialization is conserved between mouse and human lungs but is not found in alligator or turtle lungs, suggesting it arose during the evolution of the mammalian lung. The discovery of cell type specialization in alveolar capillaries transforms our understanding of the structure, function, regulation and maintenance of the air-blood barrier and gas exchange in health, disease and evolution.


Asunto(s)
Capilares/citología , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/citología , Intercambio Gaseoso Pulmonar , Envejecimiento , Caimanes y Cocodrilos/anatomía & histología , Animales , Evolución Biológica , Capilares/metabolismo , División Celular , Autorrenovación de las Células , Senescencia Celular , Humanos , Masculino , Ratones , Alveolos Pulmonares/metabolismo , Células Madre/clasificación , Células Madre/citología , Tortugas/anatomía & histología
8.
FASEB J ; 38(8): e23612, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38648494

RESUMEN

Considerable progress has been made in understanding the function of alveolar epithelial cells in a quiescent state and regeneration mechanism after lung injury. Lung injury occurs commonly from severe viral and bacterial infections, inhalation lung injury, and indirect injury sepsis. A series of pathological mechanisms caused by excessive injury, such as apoptosis, autophagy, senescence, and ferroptosis, have been studied. Recovery from lung injury requires the integrity of the alveolar epithelial cell barrier and the realization of gas exchange function. Regeneration mechanisms include the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and proteins. While alveoli are damaged, alveolar type II (AT2) cells proliferate and differentiate into alveolar type I (AT1) cells to repair the damaged alveolar epithelial layer. Alveolar epithelial cells are surrounded by various cells, such as fibroblasts, endothelial cells, and various immune cells, which affect the proliferation and differentiation of AT2 cells through paracrine during alveolar regeneration. Besides, airway epithelial cells also contribute to the repair and regeneration process of alveolar epithelium. In this review, we mainly discuss the participation of epithelial progenitor cells and various niche cells involving several signaling pathways and transcription factors.


Asunto(s)
Células Epiteliales Alveolares , Lesión Pulmonar , Regeneración , Humanos , Regeneración/fisiología , Animales , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Madre/metabolismo , Células Madre/fisiología , Alveolos Pulmonares/patología , Alveolos Pulmonares/metabolismo , Transducción de Señal , Diferenciación Celular
9.
Am J Respir Cell Mol Biol ; 70(5): 339-350, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38207121

RESUMEN

In vitro lung research requires appropriate cell culture models that adequately mimic in vivo structure and function. Previously, researchers extensively used commercially available and easily expandable A549 and NCI-H441 cells, which replicate some but not all features of alveolar epithelial cells. Specifically, these cells are often restricted by terminally altered expression while lacking important alveolar epithelial characteristics. Of late, human primary alveolar epithelial cells (hPAEpCs) have become commercially available but are so far poorly specified. Here, we applied a comprehensive set of technologies to characterize their morphology, surface marker expression, transcriptomic profile, and functional properties. At optimized seeding numbers of 7,500 cells per square centimeter and growth at a gas-liquid interface, hPAEpCs formed regular monolayers with tight junctions and amiloride-sensitive transepithelial ion transport. Electron microscopy revealed lamellar body and microvilli formation characteristic for alveolar type II cells. Protein and single-cell transcriptomic analyses revealed expression of alveolar type I and type II cell markers; yet, transcriptomic data failed to detect NKX2-1, an important transcriptional regulator of alveolar cell differentiation. With increasing passage number, hPAEpCs transdifferentiated toward alveolar-basal intermediates characterized as SFTPC-, KRT8high, and KRT5- cells. In spite of marked changes in the transcriptome as a function of passaging, Uniform Manifold Approximation and Projection plots did not reveal major shifts in cell clusters, and epithelial permeability was unaffected. The present work delineates optimized culture conditions, cellular characteristics, and functional properties of commercially available hPAEpCs. hPAEpCs may provide a useful model system for studies on drug delivery, barrier function, and transepithelial ion transport in vitro.


Asunto(s)
Células Epiteliales Alveolares , Humanos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/ultraestructura , Diferenciación Celular , Transcriptoma , Células Cultivadas , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/citología , Uniones Estrechas/metabolismo
10.
Am J Respir Cell Mol Biol ; 71(1): 30-42, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38579159

RESUMEN

Alveoli are complex microenvironments composed of various cell types, including epithelial, fibroblast, endothelial, and immune cells, which work together to maintain a delicate balance in the lung environment, ensuring proper growth, development, and an effective response to lung injuries. However, prolonged inflammation or aging can disrupt normal interactions among these cells, leading to impaired repair processes and a substantial decline in lung function. Therefore, it is essential to understand the key mechanisms underlying the interactions among the major cell types within the alveolar microenvironment. We explored the key mechanisms underlying the interactions among the major cell types within the alveolar microenvironment. These interactions occur through the secretion of signaling factors and play crucial roles in the response to injury, repair mechanisms, and the development of fibrosis in the lungs. Specifically, we focused on the regulation of alveolar type 2 cells by fibroblasts, endothelial cells, and macrophages. In addition, we explored the diverse phenotypes of fibroblasts at different stages of life and in response to lung injury, highlighting their impact on matrix production and immune functions. Furthermore, we summarize the various phenotypes of macrophages in lung injury and fibrosis as well as their intricate interplay with other cell types. This interplay can either contribute to the restoration of immune homeostasis in the alveoli or impede the repair process. Through a comprehensive exploration of these cell interactions, we aim to reveal new insights into the molecular mechanisms that drive lung injury toward fibrosis and identify potential targets for therapeutic intervention.


Asunto(s)
Comunicación Celular , Microambiente Celular , Fibroblastos , Lesión Pulmonar , Alveolos Pulmonares , Humanos , Animales , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Fibrosis , Macrófagos/metabolismo , Macrófagos/patología
11.
Pflugers Arch ; 476(7): 1155-1168, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38740599

RESUMEN

Chronic obstructive pulmonary disease (COPD) is considered a severe disease mitigating lung physiological functions with high mortality outcomes, insufficient therapy, and pathophysiology pathways which is still not fully understood. Mesenchymal stem cells (MSCs) derived from bone marrow play an important role in improving the function of organs suffering inflammation, oxidative stress, and immune reaction. It might also play a role in regenerative medicine, but that is still questionable. Additionally, Melatonin with its known antioxidative and anti-inflammatory impact is attracting attention nowadays as a useful treatment. We hypothesized that Melatonin may augment the effect of MSCs at the level of angiogenesis in COPD. In our study, the COPD model was established using cigarette smoking and lipopolysaccharide. The COPD rats were divided into four groups: COPD group, Melatonin-treated group, MSC-treated group, and combined treated group (Melatonin-MSCs). We found that COPD was accompanied by deterioration of pulmonary function tests in response to expiratory parameter affection more than inspiratory ones. This was associated with increased Hypoxia inducible factor-1α expression and vascular endothelial growth factor level. Consequently, there was increased CD31 expression indicating increased angiogenesis with massive enlargement of airspaces and thinning of alveolar septa with decreased mean radial alveolar count, in addition to, inflammatory cell infiltration and disruption of the bronchiolar epithelial wall with loss of cilia and blood vessel wall thickening. These findings were improved significantly when Melatonin and bone marrow-derived MSCs were used as a combined treatment proving the hypothesized target that Melatonin might augment MSCs aiming at vascular changes.


Asunto(s)
Melatonina , Trasplante de Células Madre Mesenquimatosas , Enfermedad Pulmonar Obstructiva Crónica , Melatonina/farmacología , Melatonina/administración & dosificación , Animales , Enfermedad Pulmonar Obstructiva Crónica/terapia , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Ratas , Masculino , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ratas Sprague-Dawley , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Angiogénesis
12.
Am J Physiol Lung Cell Mol Physiol ; 326(4): L409-L418, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38349124

RESUMEN

Alveolar type I (ATI) cells cover >95% of the lung's distal surface and facilitate gas exchange through their exceptionally thin shape. ATI cells in vivo are replenished by alveolar type II cell division and differentiation, but a detailed understanding of ATI biology has been hampered by the challenges in direct isolation of these cells due to their fragility and incomplete understanding of the signaling interactions that promote differentiation of ATII to ATI cells. Here, we explored the signals that maintain ATII versus promote ATI fates in three-dimensional (3-D) organoid cultures and developed a human alveolar type I differentiation medium (hATIDM) suitable for generating ATI cells from either mixed distal human lung cells or purified ATII cells. This media adds bone morphogenetic protein 4 (BMP4) and removes epidermal growth factor (EGF), Wnt agonist CHIR99021, and transforming growth factor-beta (TGF-ß) inhibitor SB431542 from previously developed alveolar organoid culture media. We demonstrate that BMP4 promotes expression of the ATI marker gene AGER and HOPX, whereas CHIR99021 and SB431542 maintain expression of the ATII marker gene SFTPC. The human ATI spheroids generated with hATIDM express multiple molecular and morphological features reminiscent of human ATI cells. Our results demonstrate that signaling interactions among BMP, TGF-ß, and Wnt signaling pathways in alveolar spheroids and distal lung organoids including IPF-organoids coordinate human ATII to ATI differentiation.NEW & NOTEWORTHY Alveolar type I (ATI) epithelial cells perform essential roles in maintaining lung function but have been challenging to study. We explored the signals that promote ATI fate in 3-D organoid cultures generated from either mixed distal human lung cells or purified alveolar type II (ATII) cells. This work fills an important void in our experimental repertoire for studying alveolar epithelial cells and identifies signals that promote human ATII to ATI cell differentiation.


Asunto(s)
Células Epiteliales Alveolares , Benzamidas , Dioxoles , Alveolos Pulmonares , Humanos , Alveolos Pulmonares/metabolismo , Células Cultivadas , Células Epiteliales Alveolares/metabolismo , Pulmón , Diferenciación Celular , Factor de Crecimiento Transformador beta/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L160-L172, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38771132

RESUMEN

The alveolar type II epithelial cells (AEC2s) act as stem cells in the lung for alveolar epithelial maintenance and repair. Chemokine C-X-C motif chemokine 10 (CXCL10) is expressed in injured tissues, modulating multiple cellular functions. AEC2s, previously reported to release chemokines to recruit leukocytes, were found in our study to secrete CXCL10 after bleomycin injury. We found that Sftpc-Cxcl10 transgenic mice were protected from bleomycin injury. The transgenic mice showed an increase in the AEC2 population in the lung by flow cytometry analysis. Both endogenous and exogenous CXCL10 promoted the colony formation efficiency of AEC2s in a three-dimensional (3-D) organoid growth assay. We identified that the regenerative effect of CXCL10 was CXCR3 independent using Cxcr3-deficient mice, but it was related to the TrkA pathway. Binding experiments showed that CXCL10 interacted with TrkA directly and reversibly. This study demonstrates a previously unidentified AEC2 autocrine signaling of CXCL10 to promote their regeneration and proliferation, probably involving a CXCR3-independent TrkA pathway.NEW & NOTEWORTHY CXCL10 may aid in lung injury recovery by promoting the proliferation of alveolar stem cells and using a distinct regulatory pathway from the classical one.


Asunto(s)
Células Epiteliales Alveolares , Quimiocina CXCL10 , Receptores CXCR3 , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Proliferación Celular , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptores CXCR3/metabolismo , Receptores CXCR3/genética , Regeneración , Transducción de Señal
14.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L524-L538, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38375572

RESUMEN

Lung surfactant collectins, surfactant protein A (SP-A) and D (SP-D), are oligomeric C-type lectins involved in lung immunity. Through their carbohydrate recognition domain, they recognize carbohydrates at pathogen surfaces and initiate lung innate immune response. Here, we propose that they may also be able to bind to other carbohydrates present in typical cell surfaces, such as the alveolar epithelial glycocalyx. To test this hypothesis, we analyzed and quantified the binding affinity of SP-A and SP-D to different sugars and glycosaminoglycans (GAGs) by microscale thermophoresis (MST). In addition, by changing the calcium concentration, we aimed to characterize any consequences on the binding behavior. Our results show that both oligomeric proteins bind with high affinity (in nanomolar range) to GAGs, such as hyaluronan (HA), heparan sulfate (HS) and chondroitin sulfate (CS). Binding to HS and CS was calcium-independent, as it was not affected by changing calcium concentration in the buffer. Quantification of GAGs in bronchoalveolar lavage (BAL) fluid from animals deficient in either SP-A or SP-D showed changes in GAG composition, and electron micrographs showed differences in alveolar glycocalyx ultrastructure in vivo. Taken together, SP-A and SP-D bind to model sulfated glycosaminoglycans of the alveolar epithelial glycocalyx in a multivalent and calcium-independent way. These findings provide a potential mechanism for SP-A and SP-D as an integral part of the alveolar epithelial glycocalyx binding and interconnecting free GAGs, proteoglycans, and other glycans in glycoproteins, which may influence glycocalyx composition and structure.NEW & NOTEWORTHY SP-A and SP-D function has been related to innate immunity of the lung based on their binding to sugar residues at pathogen surfaces. However, their function in the healthy alveolus was considered as limited to interaction with surfactant lipids. Here, we demonstrated that these proteins bind to glycosaminoglycans present at typical cell surfaces like the alveolar epithelial glycocalyx. We propose a model where these proteins play an important role in interconnecting alveolar epithelial glycocalyx components.


Asunto(s)
Calcio , Glicocálix , Glicosaminoglicanos , Alveolos Pulmonares , Proteína A Asociada a Surfactante Pulmonar , Proteína D Asociada a Surfactante Pulmonar , Animales , Humanos , Ratones , Células Epiteliales Alveolares/metabolismo , Líquido del Lavado Bronquioalveolar , Calcio/metabolismo , Glicocálix/metabolismo , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Ratones Endogámicos C57BL , Unión Proteica , Alveolos Pulmonares/metabolismo , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L713-L726, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38469649

RESUMEN

Mucopolysaccharidosis type IIIA (MPS IIIA) is characterized by neurological and skeletal pathologies caused by reduced activity of the lysosomal hydrolase, sulfamidase, and the subsequent primary accumulation of undegraded heparan sulfate (HS). Respiratory pathology is considered secondary in MPS IIIA and the mechanisms are not well understood. Changes in the amount, metabolism, and function of pulmonary surfactant, the substance that regulates alveolar interfacial surface tension and modulates lung compliance and elastance, have been reported in MPS IIIA mice. Here we investigated changes in lung function in 20-wk-old control and MPS IIIA mice with a closed and open thoracic cage, diaphragm contractile properties, and potential parenchymal remodeling. MPS IIIA mice had increased compliance and airway resistance and reduced tissue damping and elastance compared with control mice. The chest wall impacted lung function as observed by an increase in airway resistance and a decrease in peripheral energy dissipation in the open compared with the closed thoracic cage state in MPS IIIA mice. Diaphragm contractile forces showed a decrease in peak twitch force, maximum specific force, and the force-frequency relationship but no change in muscle fiber cross-sectional area in MPS IIIA mice compared with control mice. Design-based stereology did not reveal any parenchymal remodeling or destruction of alveolar septa in the MPS IIIA mouse lung. In conclusion, the increased storage of HS which leads to biochemical and biophysical changes in pulmonary surfactant also affects lung and diaphragm function, but has no impact on lung or diaphragm structure at this stage of the disease.NEW & NOTEWORTHY Heparan sulfate storage in the lungs of mucopolysaccharidosis type IIIA (MPS IIIA) mice leads to changes in lung function consistent with those of an obstructive lung disease and includes an increase in lung compliance and airway resistance and a decrease in tissue elastance. In addition, diaphragm muscle contractile strength is reduced, potentially further contributing to lung function impairment. However, no changes in parenchymal lung structure were observed in mice at 20 wk of age.


Asunto(s)
Resistencia de las Vías Respiratorias , Diafragma , Mucopolisacaridosis III , Alveolos Pulmonares , Animales , Diafragma/fisiopatología , Diafragma/patología , Diafragma/metabolismo , Rendimiento Pulmonar , Ratones , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Alveolos Pulmonares/metabolismo , Mucopolisacaridosis III/patología , Mucopolisacaridosis III/fisiopatología , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/genética , Contracción Muscular/fisiología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Fuerza Muscular , Pulmón/patología , Pulmón/fisiopatología , Pulmón/metabolismo , Masculino
16.
Biochem Biophys Res Commun ; 727: 150308, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38968769

RESUMEN

Excessive autophagy may lead to degradation and damage of alveolar epithelial cells after lung transplantation, eventually leading to alveolar epithelial cell loss, affecting the structural integrity and function of alveoli. Glutamine (Gln), a nutritional supplement, regulates autophagy through multiple signaling pathways. In this study, we explored the protective role of Gln on alveolar epithelial cells by inhibiting autophagy. In vivo, a rat orthotopic lung transplant model was carried out to evaluate the therapeutic effect of glutamine. Ischemia/reperfusion (I/R) induced alveolar collapse, edema, epithelial cell apoptosis, and inflammation, which led to a reduction of alveolar physiological function, such as an increase in peak airway pressure, and a decrease in lung compliance and oxygenation index. In comparison, Gln preserved alveolar structure and function by reducing alveolar apoptosis, inflammation, and edema. In vitro, a hypoxia/reoxygenation (H/R) cell model was performed to simulate IR injury on mouse lung epithelial (MLE) cells and human lung bronchus epithelial (Beas-2B) cells. H/R impaired the proliferation of epithelial cells and triggered cell apoptosis. In contrast, Gln normalized cell proliferation and suppressed I/R-induced cell apoptosis. The activation of mTOR and the downregulation of autophagy-related proteins (LC3, Atg5, Beclin1) were observed in Gln-treated lung tissues and alveolar epithelial cells. Both in vivo and in vitro, rapamycin, a classical mTOR inhibitor, reversed the beneficial effects of Gln on alveolar structure and function. Taken together, Glnpreserved alveolar structure and function after lung transplantation by inhibiting autophagy.


Asunto(s)
Autofagia , Glutamina , Trasplante de Pulmón , Alveolos Pulmonares , Ratas Sprague-Dawley , Daño por Reperfusión , Autofagia/efectos de los fármacos , Animales , Glutamina/metabolismo , Glutamina/farmacología , Masculino , Humanos , Ratones , Ratas , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Apoptosis/efectos de los fármacos , Línea Celular , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología
17.
Part Fibre Toxicol ; 21(1): 25, 2024 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760786

RESUMEN

Exposure to indoor air pollutants (IAP) has increased recently, with people spending more time indoors (i.e. homes, offices, schools and transportation). Increased exposures of IAP on a healthy population are poorly understood, and those with allergic respiratory conditions even less so. The objective of this study, therefore, was to implement a well-characterised in vitro model of the human alveolar epithelial barrier (A549 + PMA differentiated THP-1 incubated with and without IL-13, IL-5 and IL-4) to determine the effects of a standardised indoor particulate (NIST 2583) on both a healthy lung model and one modelling a type-II (stimulated with IL-13, IL-5 and IL-4) inflammatory response (such as asthma).Using concentrations from the literature, and an environmentally appropriate exposure we investigated 232, 464 and 608ng/cm2 of NIST 2583 respectively. Membrane integrity (blue dextran), viability (trypan blue), genotoxicity (micronucleus (Mn) assay) and (pro-)/(anti-)inflammatory effects (IL-6, IL-8, IL-33, IL-10) were then assessed 24 h post exposure to both models. Models were exposed using a physiologically relevant aerosolisation method (VitroCell Cloud 12 exposure system).No changes in Mn frequency or membrane integrity in either model were noted when exposed to any of the tested concentrations of NIST 2583. A significant decrease (p < 0.05) in cell viability at the highest concentration was observed in the healthy model. Whilst cell viability in the "inflamed" model was decreased at the lower concentrations (significantly (p < 0.05) after 464ng/cm2). A significant reduction (p < 0.05) in IL-10 and a significant increase in IL-33 was seen after 24 h exposure to NIST 2583 (464, 608ng/cm2) in the "inflamed" model.Collectively, the results indicate the potential for IAP to cause the onset of a type II response as well as exacerbating pre-existing allergic conditions. Furthermore, the data imposes the importance of considering unhealthy individuals when investigating the potential health effects of IAP. It also highlights that even in a healthy population these particles have the potential to induce this type II response and initiate an immune response following exposure to IAP.


Asunto(s)
Contaminación del Aire Interior , Supervivencia Celular , Material Particulado , Humanos , Contaminación del Aire Interior/efectos adversos , Material Particulado/toxicidad , Supervivencia Celular/efectos de los fármacos , Células A549 , Citocinas/metabolismo , Células THP-1 , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Contaminantes Atmosféricos/toxicidad , Inflamación/inducido químicamente , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología
18.
Part Fibre Toxicol ; 21(1): 26, 2024 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778339

RESUMEN

BACKGROUND: During inhalation, airborne particles such as particulate matter ≤ 2.5 µm (PM2.5), can deposit and accumulate on the alveolar epithelial tissue. In vivo studies have shown that fractions of PM2.5 can cross the alveolar epithelium to blood circulation, reaching secondary organs beyond the lungs. However, approaches to quantify the translocation of particles across the alveolar epithelium in vivo and in vitro are still not well established. In this study, methods to assess the translocation of standard diesel exhaust particles (DEPs) across permeable polyethylene terephthalate (PET) inserts at 0.4, 1, and 3 µm pore sizes were first optimized with transmission electron microscopy (TEM), ultraviolet-visible spectroscopy (UV-VIS), and lock-in thermography (LIT), which were then applied to study the translocation of DEPs across human alveolar epithelial type II (A549) cells. A549 cells that grew on the membrane (pore size: 3 µm) in inserts were exposed to DEPs at different concentrations from 0 to 80 µg.mL- 1 ( 0 to 44 µg.cm- 2) for 24 h. After exposure, the basal fraction was collected and then analyzed by combining qualitative (TEM) and quantitative (UV-VIS and LIT) techniques to assess the translocated fraction of the DEPs across the alveolar epithelium in vitro. RESULTS: We could detect the translocated fraction of DEPs across the PET membranes with 3 µm pore sizes and without cells by TEM analysis, and determine the percentage of translocation at approximatively 37% by UV-VIS (LOD: 1.92 µg.mL- 1) and 75% by LIT (LOD: 0.20 µg.cm- 2). In the presence of cells, the percentage of DEPs translocation across the alveolar tissue was determined around 1% at 20 and 40 µg.mL- 1 (11 and 22 µg.cm- 2), and no particles were detected at higher and lower concentrations. Interestingly, simultaneous exposure of A549 cells to DEPs and EDTA can increase the translocation of DEPs in the basal fraction. CONCLUSION: We propose a combination of analytical techniques to assess the translocation of DEPs across lung tissues. Our results reveal a low percentage of translocation of DEPs across alveolar epithelial tissue in vitro and they correspond to in vivo findings. The combination approach can be applied to any traffic-generated particles, thus enabling us to understand their involvement in public health.


Asunto(s)
Material Particulado , Alveolos Pulmonares , Emisiones de Vehículos , Humanos , Emisiones de Vehículos/toxicidad , Emisiones de Vehículos/análisis , Células A549 , Material Particulado/toxicidad , Material Particulado/análisis , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Tamaño de la Partícula , Microscopía Electrónica de Transmisión , Tereftalatos Polietilenos/química , Tereftalatos Polietilenos/toxicidad , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/análisis
19.
Cell Mol Life Sci ; 80(6): 145, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37166489

RESUMEN

Alveolar epithelial type II cells (AT2s) together with AT1s constitute the epithelial lining of lung alveoli. In contrast to the large flat AT1s, AT2s are cuboidal and smaller. In addition to surfactant production, AT2s also serve as prime alveolar progenitors in homeostasis and play an important role during regeneration/repair. Based on different lineage tracing strategies in mice and single-cell transcriptomic analysis, recent reports highlight the heterogeneous nature of AT2s. These studies present compelling evidence for the presence of stable or transitory AT2 subpopulations with distinct marker expression, signaling pathway activation and functional properties. Despite demonstrated progenitor potentials of AT2s in maintaining homeostasis, through self-renewal and differentiation to AT1s, the exact identity, full progenitor potential and regulation of these progenitor cells, especially in the context of human diseases remain unclear. We recently identified a novel subset of AT2 progenitors named "Injury-Activated Alveolar Progenitors" (IAAPs), which express low levels of Sftpc, Sftpb, Sftpa1, Fgfr2b and Etv5, but are highly enriched for the expression of the surface receptor programmed cell death-ligand 1 (Pd-l1). IAAPs are quiescent during lung homeostasis but activated upon injury with the potential to proliferate and differentiate into AT2s. Significantly, a similar population of PD-L1 positive cells expressing intermediate levels of SFTPC are found to be expanded in human IPF lungs. We summarize here the current understanding of this newly discovered AT2 progenitor subpopulation and also try to reconcile the relationship between different AT2 stem cell subpopulations regarding their progenitor potential, regulation, and relevance to disease pathogenesis and therapeutic interventions.


Asunto(s)
Antígeno B7-H1 , Pulmón , Ratones , Humanos , Animales , Antígeno B7-H1/metabolismo , Pulmón/metabolismo , Células Epiteliales Alveolares , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Diferenciación Celular/fisiología
20.
Int J Clin Oncol ; 29(6): 771-779, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38600426

RESUMEN

BACKGROUND: Adenocarcinomas show a stepwise progression from atypical adenomatous hyperplasia (AAH) through adenocarcinoma in situ (AIS) to invasive adenocarcinoma (IA). Immunoglobulin superfamily containing leucine-rich repeat (ISLR) is a marker of tumor-restraining cancer-associated fibroblasts (CAFs), which are distinct from conventional, strongly α-smooth muscle actin (αSMA)-positive CAFs. Fibroblast activation protein (FAP) has been focused on as a potential therapeutic and diagnostic target of CAFs. METHODS: We investigated the changes in protein expression during adenocarcinoma progression in the pre-existing alveolar septa by assessing ISLR, αSMA, and FAP expression in normal lung, AAH, AIS, and IA. Fourteen AAH, seventeen AIS, and twenty IA lesions were identified and randomly sampled. Immunohistochemical analysis was performed to evaluate cancer-associated changes and FAP expression in the pre-existing alveolar structures. RESULTS: Normal alveolar septa expressed ISLR. The ISLR level in the alveolar septa decreased in AAH and AIS tissues when compared with that in normal lung tissue. The αSMA-positive area gradually increased from the adjacent lung tissue (13.3% ± 15%) to AIS (87.7% ± 14%), through AAH (70.2% ± 21%). Moreover, the FAP-positive area gradually increased from AAH (1.69% ± 1.4%) to IA (11.8% ± 7.1%), through AIS (6.11% ± 5.3%). Protein expression changes are a feature of CAFs in the pre-existing alveolar septa that begin in AAH. These changes gradually progressed from AAH to IA through AIS. CONCLUSIONS: FAP-positive fibroblasts may contribute to tumor stroma formation in early-stage lung adenocarcinoma, and this could influence the development of therapeutic strategies targeting FAP-positive CAFs for disrupting extracellular matrix formation.


Asunto(s)
Adenocarcinoma del Pulmón , Progresión de la Enfermedad , Endopeptidasas , Neoplasias Pulmonares , Proteínas de la Membrana , Humanos , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Proteínas de la Membrana/metabolismo , Anciano , Gelatinasas/metabolismo , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/genética , Actinas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Biomarcadores de Tumor/metabolismo , Alveolos Pulmonares/patología , Alveolos Pulmonares/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Estadificación de Neoplasias , Adenocarcinoma in Situ/patología , Adenocarcinoma in Situ/metabolismo , Adulto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA