Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Eur Respir J ; 63(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609098

RESUMEN

Electronic cigarette (e-cigarette) use continues to rise globally. E-cigarettes have been presented as safer alternatives to combustion cigarettes that can mitigate the harm associated with tobacco products; however, the degree to which e-cigarette use itself can lead to morbidity and mortality is not fully defined. Herein we describe how e-cigarettes function; discuss the current knowledge of the effects of e-cigarette aerosol on lung cell cytotoxicity, inflammation, antipathogen immune response, mucociliary clearance, oxidative stress, DNA damage, carcinogenesis, matrix remodelling and airway hyperresponsiveness; and summarise the impact on lung diseases, including COPD, respiratory infection, lung cancer and asthma. We highlight how the inclusion of nicotine or flavouring compounds in e-liquids can impact lung toxicity. Finally, we consider the paradox of the safer cigarette: the toxicities of e-cigarettes that can mitigate their potential to serve as a harm reduction tool in the fight against traditional cigarettes, and we summarise the research needed in this underinvestigated area.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Pulmón , Humanos , Pulmón/efectos de los fármacos , Enfermedades Pulmonares/inducido químicamente , Nicotina/efectos adversos , Reducción del Daño , Estrés Oxidativo , Vapeo/efectos adversos , Daño del ADN , Productos de Tabaco/efectos adversos
2.
Eur Respir J ; 63(1)2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37918852

RESUMEN

RATIONALE: Recent data suggest that the localisation of airway epithelial cells in the distal lung in idiopathic pulmonary fibrosis (IPF) may drive pathology. We set out to discover whether chemokines expressed in these ectopic airway epithelial cells may contribute to the pathogenesis of IPF. METHODS: We analysed whole lung and single-cell transcriptomic data obtained from patients with IPF. In addition, we measured chemokine levels in blood, bronchoalveolar lavage (BAL) of IPF patients and air-liquid interface cultures. We employed ex vivo donor and IPF lung fibroblasts and an animal model of pulmonary fibrosis to test the effects of chemokine signalling on fibroblast function. RESULTS: By analysis of whole-lung transcriptomics, protein and BAL, we discovered that CXCL6 (a member of the interleukin-8 family) was increased in patients with IPF. Elevated CXCL6 levels in the BAL of two cohorts of patients with IPF were associated with poor survival (hazard ratio of death or progression 1.89, 95% CI 1.16-3.08; n=179, p=0.01). By immunostaining and single-cell RNA sequencing, CXCL6 was detected in secretory cells. Administration of mCXCL5 (LIX, murine CXCL6 homologue) to mice increased collagen synthesis with and without bleomycin. CXCL6 increased collagen I levels in donor and IPF fibroblasts 4.4-fold and 1.7-fold, respectively. Both silencing of and chemical inhibition of CXCR1/2 blocked the effects of CXCL6 on collagen, while overexpression of CXCR2 increased collagen I levels 4.5-fold in IPF fibroblasts. CONCLUSIONS: CXCL6 is expressed in ectopic airway epithelial cells. Elevated levels of CXCL6 are associated with IPF mortality. CXCL6-driven collagen synthesis represents a functional consequence of ectopic localisation of airway epithelial cells in IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Humanos , Ratones , Bleomicina , Quimiocina CXCL6/metabolismo , Quimiocinas/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/genética , Pulmón/patología
3.
Am J Respir Crit Care Med ; 209(4): 362-373, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38113442

RESUMEN

Despite progress in elucidation of disease mechanisms, identification of risk factors, biomarker discovery, and the approval of two medications to slow lung function decline in idiopathic pulmonary fibrosis and one medication to slow lung function decline in progressive pulmonary fibrosis, pulmonary fibrosis remains a disease with a high morbidity and mortality. In recognition of the need to catalyze ongoing advances and collaboration in the field of pulmonary fibrosis, the NHLBI, the Three Lakes Foundation, and the Pulmonary Fibrosis Foundation hosted the Pulmonary Fibrosis Stakeholder Summit on November 8-9, 2022. This workshop was held virtually and was organized into three topic areas: 1) novel models and research tools to better study pulmonary fibrosis and uncover new therapies, 2) early disease risk factors and methods to improve diagnosis, and 3) innovative approaches toward clinical trial design for pulmonary fibrosis. In this workshop report, we summarize the content of the presentations and discussions, enumerating research opportunities for advancing our understanding of the pathogenesis, treatment, and outcomes of pulmonary fibrosis.


Asunto(s)
Investigación Biomédica , Fibrosis Pulmonar Idiopática , Estados Unidos , Humanos , National Heart, Lung, and Blood Institute (U.S.) , Lagos , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/terapia , Factores de Riesgo
4.
Am J Respir Cell Mol Biol ; 69(5): 556-569, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37487137

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by progressive lung scarring and remodeling. Although treatments exist that slow disease progression, IPF is irreversible, and there is no cure. Cellular senescence, a major hallmark of aging, has been implicated in IPF pathogenesis, and mitochondrial dysfunction is increasingly recognized as a driver of senescence. Adenine nucleotide translocases (ANTs) are abundant mitochondrial ATP-ADP transporters critical for regulating cell fate and maintaining mitochondrial function. We sought to determine how alterations in ANTs influence cellular senescence in pulmonary fibrosis. We found that SLC25A4 (solute carrier family 25 member 4) (ANT1) and SLC25A5 (ANT2) expression is reduced in the lungs of patients with IPF, particularly within alveolar type II (AT2) cells, by single-cell RNA sequencing and tissue staining. Loss of ANT1 by siRNA in lung epithelial cells resulted in increased senescence markers such as ß-galactosidase and p21, with a reduction in the ratio of nicotinamide adenine dinucleotide to reduced nicotinamide adenine dinucleotide. Bleomycin-treated ANT1 knockdown cells also had increased senescence markers compared with bleomycin-treated control cells. Loss of ANT1 in AT2 cells resulted in a reduction in alveolar organoid growth, with an increase in p21 by staining. Global loss of ANT1 resulted in worse lung fibrosis and increased senescence in the bleomycin- and asbestos-induced mouse models of pulmonary fibrosis. In summary, loss of ANT1 contributes to IPF pathogenesis through mitochondrial dysfunction, increased senescence, and decreased regenerative capacity of AT2 cells, resulting in enhanced lung fibrosis. Modulation of ANTs presents a new therapeutic avenue that may alter cellular senescence pathways and limit pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática , NAD , Animales , Humanos , Ratones , Bleomicina/farmacología , Senescencia Celular , Células Epiteliales/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , NAD/metabolismo
5.
Dev Biol ; 501: 111-123, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37353105

RESUMEN

Smoking cigarettes during pregnancy is associated with adverse effects on infants including low birth weight, defective lung development, and skeletal abnormalities. Pregnant women are increasingly turning to vaping [use of electronic (e)-cigarettes] as a perceived safer alternative to cigarettes. However, nicotine disrupts fetal development, suggesting that like cigarette smoking, nicotine vaping may be detrimental to the fetus. To test the impact of maternal vaping on fetal lung and skeletal development in mice, pregnant dams were exposed to e-cigarette vapor throughout gestation. At embryonic day (E)18.5, vape exposed litter sizes were reduced, and some embryos exhibited growth restriction compared to air exposed controls. Fetal lungs were collected for histology and whole transcriptome sequencing. Maternally nicotine vaped embryos exhibited histological and transcriptional changes consistent with impaired distal lung development. Embryonic lung gene expression changes mimicked transcriptional changes observed in adult mouse lungs exposed to cigarette smoke, suggesting that the developmental defects may be due to direct nicotine exposure. Fetal skeletons were analyzed for craniofacial and long bone lengths. Nicotine directly binds and inhibits the Kcnj2 potassium channel which is important for bone development. The length of the maxilla, palatal shelves, humerus, and femur were reduced in vaped embryos, which was further exacerbated by loss of one copy of the Kcnj2 gene. Nicotine vapor exposed Kcnj2KO/+ embryos also had significantly lower birth weights than unexposed animals of either genotype. Kcnj2 mutants had severely defective lungs with and without vape exposure, suggesting that potassium channels may be broadly involved in mediating the detrimental developmental effects of nicotine vaping. These data indicate that intrauterine nicotine exposure disrupts fetal lung and skeletal development likely through inhibition of Kcnj2.


Asunto(s)
Cigarrillo Electrónico a Vapor , Sistemas Electrónicos de Liberación de Nicotina , Vapeo , Femenino , Embarazo , Animales , Humanos , Ratones , Vapeo/efectos adversos , Nicotina/efectos adversos , Nicotina/metabolismo , Pulmón/metabolismo , Cigarrillo Electrónico a Vapor/efectos adversos
6.
Am J Respir Cell Mol Biol ; 69(2): 126-134, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37236629

RESUMEN

Chord length is an indirect measure of alveolar size and a critical endpoint in animal models of chronic obstructive pulmonary disease (COPD). In assessing chord length, the lumens of nonalveolar structures are eliminated from measurement by various methods, including manual masking. However, manual masking is resource intensive and can introduce variability and bias. We created a fully automated deep learning-based tool to mask murine lung images and assess chord length to facilitate mechanistic and therapeutic discovery in COPD called Deep-Masker (available at http://47.93.0.75:8110/login). We trained the deep learning algorithm for Deep-Masker using 1,217 images from 137 mice from 12 strains exposed to room air or cigarette smoke for 6 months. We validated this algorithm against manual masking. Deep-Masker demonstrated high accuracy with an average difference in chord length compared with manual masking of -0.3 ± 1.4% (rs = 0.99) for room-air-exposed mice and 0.7 ± 1.9% (rs = 0.99) for cigarette-smoke-exposed mice. The difference between Deep-Masker and manually masked images for change in chord length because of cigarette smoke exposure was 6.0 ± 9.2% (rs = 0.95). These values exceed published estimates for interobserver variability for manual masking (rs = 0.65) and the accuracy of published algorithms by a significant margin. We validated the performance of Deep-Masker using an independent set of images. Deep-Masker can be an accurate, precise, fully automated method to standardize chord length measurement in murine models of lung disease.


Asunto(s)
Aprendizaje Profundo , Enfermedad Pulmonar Obstructiva Crónica , Animales , Ratones , Pulmón , Enfermedad Pulmonar Obstructiva Crónica/diagnóstico por imagen
7.
Cell Rep ; 42(6): 112533, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37257449

RESUMEN

The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Molécula de Adhesión Celular Epitelial , Neoplasias de la Mama/patología , Línea Celular Tumoral , Mama/metabolismo , Células Clonales/metabolismo , Transición Epitelial-Mesenquimal
8.
Am J Respir Cell Mol Biol ; 68(4): 444-455, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36608844

RESUMEN

Chronic obstructive pulmonary disease (COPD) is characterized by a persistent inflammatory state in the lungs and defective tissue repair. Although the inflammatory response in patients with COPD is well characterized and known to be exaggerated during exacerbations, its contribution to lung injury and abnormal repair is still unclear. In this study, we aimed to investigate how the inflammatory microenvironment affects the epithelial progenitors and their supporting mesenchymal niche cells involved in tissue repair of the distal lung. We focused on IL-1ß, a key inflammatory mediator that is increased during exacerbations of COPD, and used an organoid model of lung epithelial cells and fibroblasts to assess the effect of IL-1ß treatment on these cells' transcriptome and secreted factors. Whereas direct treatment of the lung organoids with IL-1ß promoted organoid growth, this switched toward inhibition when it was added as fibroblast pretreatment followed by organoid treatment. We then investigated the IL-1ß-driven mechanisms in the fibroblasts and found an inflammatory response related to (C-X-C motif) ligand (CXCL) chemokines; we confirmed that these chemokines were responsible for the impaired organoid growth and found that targeting their C-X-C chemokine receptors 1/2 (CXCR1/2) receptors or the IL-1ß intracellular signaling reduced the proinflammatory response and restored organoid growth. These data demonstrate that IL-1ß alters the fibroblasts' state by promoting a distinct inflammatory response, switching their supportive function on epithelial progenitors toward an inhibitory one in an organoid assay. These results imply that chronic inflammation functions as a shift toward inhibition of repair, thereby contributing to chronic inflammatory diseases like COPD.


Asunto(s)
Interleucina-1beta , Pulmón , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Enfermedad Crónica , Fibroblastos , Transducción de Señal , Interleucina-1beta/farmacología , Células Cultivadas , Células Epiteliales
10.
Am J Respir Crit Care Med ; 207(3): 283-299, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36047984

RESUMEN

Rationale: Although type II alveolar epithelial cells (AEC2s) are chronically injured in idiopathic pulmonary fibrosis (IPF), they contribute to epithelial regeneration in IPF. Objectives: We hypothesized that Notch signaling may contribute to AEC2 proliferation, dedifferentiation characterized by loss of surfactant processing machinery, and lung fibrosis in IPF. Methods: We applied microarray analysis, kinome profiling, flow cytometry, immunofluorescence analysis, western blotting, quantitative PCR, and proliferation and surface activity analysis to study epithelial differentiation, proliferation, and matrix deposition in vitro (AEC2 lines, primary murine/human AEC2s), ex vivo (human IPF-derived precision-cut lung slices), and in vivo (bleomycin and pepstatin application, Notch1 [Notch receptor 1] intracellular domain overexpression). Measurements and Main Results: We document here extensive SP-B and -C (surfactant protein-B and -C) processing defects in IPF AEC2s, due to loss of Napsin A, resulting in increased intra-alveolar surface tension and alveolar collapse and induction of endoplasmic reticulum stress in AEC2s. In vivo pharmacological inhibition of Napsin A results in the development of AEC2 injury and overt lung fibrosis. We also demonstrate that Notch1 signaling is already activated early in IPF and determines AEC2 fate by inhibiting differentiation (reduced lamellar body compartment, reduced capacity to process hydrophobic SP) and by causing increased epithelial proliferation and development of lung fibrosis, putatively via altered JAK (Janus kinase)/Stat (signal transducer and activator of transcription) signaling in AEC2s. Conversely, inhibition of Notch signaling in IPF-derived precision-cut lung slices improved the surfactant processing capacity of AEC2s and reversed fibrosis. Conclusions: Notch1 is a central regulator of AEC2 fate in IPF. It induces alveolar epithelial proliferation and loss of Napsin A and of surfactant proprotein processing, and it contributes to fibroproliferation.


Asunto(s)
Fibrosis Pulmonar Idiopática , Surfactantes Pulmonares , Humanos , Ratones , Animales , Tensoactivos , Pulmón , Células Epiteliales Alveolares , Bleomicina , Receptor Notch1
12.
Environ Pollut ; 305: 119292, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35439594

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by inflammation and impaired tissue regeneration, and is reported as the fourth leading cause of death worldwide by the Centers for Disease Control and Prevention (CDC). Environmental pollution and specifically motor vehicle emissions are known to play a role in the pathogenesis of COPD, but little is still known about the molecular mechanisms that are altered following diesel exhaust particles (DEP) exposure. Here we used lung organoids derived from co-culture of alveolar epithelial progenitors and fibroblasts to investigate the effect of DEP on the epithelial-mesenchymal signaling niche in the distal lung, which is essential for tissue repair. We found that DEP treatment impaired the number as well as the average diameter of both airway and alveolar type of lung organoids. Bulk RNA-sequencing of re-sorted epithelial cells and fibroblasts following organoid co-culture shows that the Nrf2 pathway, which regulates antioxidants' activity, was upregulated in both cell populations in response to DEP; and WNT/ß-catenin signaling, which is essential to promote epithelial repair, was downregulated in DEP-exposed epithelial cells. We show that pharmacological treatment with anti-oxidant agents such as N-acetyl cysteine (NAC) or Mitoquinone mesylate (MitoQ) reversed the effect of DEP on organoids growth. Additionally, a WNT/ß-catenin activator (CHIR99021) successfully restored WNT signaling and promoted organoid growth upon DEP exposure. We propose that targeting oxidative stress and specific signaling pathways affected by DEP in the distal lung may represent a strategy to restore tissue repair in COPD.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , beta Catenina , Células Epiteliales , Fibroblastos/patología , Humanos , Pulmón/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Emisiones de Vehículos/toxicidad , beta Catenina/metabolismo
13.
Sci Adv ; 8(12): eabj9949, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35319981

RESUMEN

Currently, there is no pharmacological treatment targeting defective tissue repair in chronic disease. Here, we used a transcriptomics-guided drug target discovery strategy using gene signatures of smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke, identifying druggable targets expressed in alveolar epithelial progenitors, of which we screened the function in lung organoids. We found several drug targets with regenerative potential, of which EP and IP prostanoid receptor ligands had the most profound therapeutic potential in restoring cigarette smoke-induced defects in alveolar epithelial progenitors in vitro and in vivo. Mechanistically, we found, using single-cell RNA sequencing analysis, that circadian clock and cell cycle/apoptosis signaling pathways were differentially expressed in alveolar epithelial progenitor cells in patients with COPD and in a relevant model of COPD, which was prevented by prostaglandin E2 or prostacyclin mimetics. We conclude that specific targeting of EP and IP receptors offers therapeutic potential for injury to repair in COPD.


Asunto(s)
Enfermedad Pulmonar Obstructiva Crónica , Transcriptoma , Animales , Humanos , Ligandos , Pulmón/metabolismo , Ratones , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/genética , Regeneración
14.
Handb Exp Pharmacol ; 269: 305-336, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34463851

RESUMEN

The main physiological function of the lung is gas exchange, mediated at the interface between the alveoli and the pulmonary microcapillary network and facilitated by conducting airway structures that regulate the transport of these gases from and to the alveoli. Exposure to microbial and environmental factors such as allergens, viruses, air pollution, and smoke contributes to the development of chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and lung cancer. Respiratory diseases as a cluster are the commonest cause of chronic disease and of hospitalization in children and are among the three most common causes of morbidity and mortality in the adult population worldwide. Many of these chronic respiratory diseases are associated with inflammation and structural remodelling of the airways and/or alveolar tissues. They can often only be treated symptomatically with no disease-modifying therapies that normalize the pathological tissue destruction driven by inflammation and remodelling. In search for novel therapeutic strategies for these diseases, several lines of evidence revealed the WNT pathway as an emerging target for regenerative strategies in the lung. WNT proteins, their receptors, and signalling effectors have central regulatory roles under (patho)physiological conditions underpinning lung function and (chronic) lung diseases and we summarize these roles and discuss how pharmacological targeting of the WNT pathway may be utilized for the treatment of chronic lung diseases.


Asunto(s)
Enfermedades Pulmonares , Pulmón/fisiología , Vía de Señalización Wnt , Adulto , Niño , Humanos , Enfermedades Pulmonares/etiología , Enfermedad Pulmonar Obstructiva Crónica , Proteínas Wnt
15.
Clin Chest Med ; 42(2): 365-373, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34024411

RESUMEN

Current therapeutic strategies have succeeded in slowing down the progression of idiopathic pulmonary fibrosis (IPF). Emerging evidence highlights IPF as a disease of aging and impaired regeneration. Novel antiaging and regenerative medicine approaches hold promise to be able to reverse disease and might present hope for a cure. Research focusing on a deeper understanding of lung stem cell populations and how these are regulated and altered in fibrotic disease continues to drive the field, and accompanied by earlier diagnosis, the adaptation of clinically relevant models and readouts for regeneration of diseased lung, ultimately paves the way for translation into clinics.


Asunto(s)
Fibrosis Pulmonar Idiopática/terapia , Medicina Regenerativa , Humanos , Pulmón , Células Madre
16.
Nature ; 588(7836): 151-156, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33149305

RESUMEN

Lymphotoxin ß-receptor (LTßR) signalling promotes lymphoid neogenesis and the development of tertiary lymphoid structures1,2, which are associated with severe chronic inflammatory diseases that span several organ systems3-6. How LTßR signalling drives chronic tissue damage particularly in the lung, the mechanism(s) that regulate this process, and whether LTßR blockade might be of therapeutic value have remained unclear. Here we demonstrate increased expression of LTßR ligands in adaptive and innate immune cells, enhanced non-canonical NF-κB signalling, and enriched LTßR target gene expression in lung epithelial cells from patients with smoking-associated chronic obstructive pulmonary disease (COPD) and from mice chronically exposed to cigarette smoke. Therapeutic inhibition of LTßR signalling in young and aged mice disrupted smoking-related inducible bronchus-associated lymphoid tissue, induced regeneration of lung tissue, and reverted airway fibrosis and systemic muscle wasting. Mechanistically, blockade of LTßR signalling dampened epithelial non-canonical activation of NF-κB, reduced TGFß signalling in airways, and induced regeneration by preventing epithelial cell death and activating WNT/ß-catenin signalling in alveolar epithelial progenitor cells. These findings suggest that inhibition of LTßR signalling represents a viable therapeutic option that combines prevention of tertiary lymphoid structures1 and inhibition of apoptosis with tissue-regenerative strategies.


Asunto(s)
Pulmón/efectos de los fármacos , Pulmón/fisiología , Receptor beta de Linfotoxina/antagonistas & inhibidores , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Wnt/agonistas , Inmunidad Adaptativa , Envejecimiento/metabolismo , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Apoptosis/efectos de los fármacos , Enfisema/metabolismo , Femenino , Humanos , Inmunidad Innata , Pulmón/metabolismo , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Humo/efectos adversos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
17.
Nat Commun ; 11(1): 3559, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32678092

RESUMEN

The cell type specific sequences of transcriptional programs during lung regeneration have remained elusive. Using time-series single cell RNA-seq of the bleomycin lung injury model, we resolved transcriptional dynamics for 28 cell types. Trajectory modeling together with lineage tracing revealed that airway and alveolar stem cells converge on a unique Krt8 + transitional stem cell state during alveolar regeneration. These cells have squamous morphology, feature p53 and NFkB activation and display transcriptional features of cellular senescence. The Krt8+ state appears in several independent models of lung injury and persists in human lung fibrosis, creating a distinct cell-cell communication network with mesenchyme and macrophages during repair. We generated a model of gene regulatory programs leading to Krt8+ transitional cells and their terminal differentiation to alveolar type-1 cells. We propose that in lung fibrosis, perturbed molecular checkpoints on the way to terminal differentiation can cause aberrant persistence of regenerative intermediate stem cell states.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Queratina-8/metabolismo , Alveolos Pulmonares/fisiología , Fibrosis Pulmonar/patología , Regeneración , Células Madre/metabolismo , Células Epiteliales Alveolares/citología , Animales , Comunicación Celular , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Queratina-8/genética , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Fibrosis Pulmonar/metabolismo , Análisis de la Célula Individual , Células Madre/citología
18.
Cell Signal ; 70: 109588, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32109549

RESUMEN

The rapid expansion of the elderly population has led to the recent epidemic of age-related diseases, including increased incidence and mortality of chronic lung diseases, such as Idiopathic Pulmonary Fibrosis (IPF). Cellular senescence is a major hallmark of aging and has a higher occurrence in IPF. The lung epithelium represents a major site of tissue injury, cellular senescence and aberrant activity of developmental pathways such as the WNT/ß-catenin pathway in IPF. The potential impact of WNT/ß-catenin signaling on alveolar epithelial senescence in general as well as in IPF, however, remains elusive. Here, we characterized alveolar epithelial cells of aged mice and assessed the contribution of chronic WNT/ß-catenin signaling on alveolar epithelial type (AT) II cell senescence. Whole lungs from old (16-24 months) versus young (3 months) mice had relatively less epithelial (EpCAM+) but more inflammatory (CD45+) cells, as assessed by flow cytometry. Compared to young ATII cells, old ATII cells showed decreased expression of the ATII cell marker Surfactant Protein C along with increased expression of the ATI cell marker Hopx, accompanied by increased WNT/ß-catenin activity. Notably, when placed in an organoid assay, old ATII cells exhibited decreased progenitor cell potential. Chronic canonical WNT/ß-catenin activation for up to 7 days in primary ATII cells as well as alveolar epithelial cell lines induced a robust cellular senescence, whereas the non-canonical ligand WNT5A was not able to induce cellular senescence. Moreover, chronic WNT3A treatment of precision-cut lung slices (PCLS) further confirmed ATII cell senescence. Simultaneously, chronic but not acute WNT/ß-catenin activation induced a profibrotic state with increased expression of the impaired ATII cell marker Keratin 8. These results suggest that chronic WNT/ß-catenin activity in the IPF lung contributes to increased ATII cell senescence and reprogramming. In the fibrotic environment, WNT/ß-catenin signaling thus might lead to further progenitor cell dysfunction and impaired lung repair.


Asunto(s)
Células Epiteliales Alveolares , Senescencia Celular , Fibrosis Pulmonar Idiopática/metabolismo , Proteínas Wnt/fisiología , Vía de Señalización Wnt , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Línea Celular , Humanos , Pulmón/citología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL
19.
Biochem Soc Trans ; 48(1): 231-243, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32096543

RESUMEN

The planar cell polarity (PCP) signaling pathway is a potent developmental regulator of directional cell behaviors such as migration, asymmetric division and morphological polarization that are critical for shaping the body axis and the complex three-dimensional architecture of tissues and organs. PCP is considered a noncanonical Wnt pathway due to the involvement of Wnt ligands and Frizzled family receptors in the absence of the beta-catenin driven gene expression observed in the canonical Wnt cascade. At the heart of the PCP mechanism are protein complexes capable of generating molecular asymmetries within cells along a tissue-wide axis that are translated into polarized actin and microtubule cytoskeletal dynamics. PCP has emerged as an important regulator of developmental, homeostatic and disease processes in the respiratory system. It acts along other signaling pathways to create the elaborately branched structure of the lung by controlling the directional protrusive movements of cells during branching morphogenesis. PCP operates in the airway epithelium to establish and maintain the orientation of respiratory cilia along the airway axis for anatomically directed mucociliary clearance. It also regulates the establishment of the pulmonary vasculature. In adult tissues, PCP dysfunction has been linked to a variety of chronic lung diseases such as cystic fibrosis, chronic obstructive pulmonary disease, and idiopathic pulmonary arterial hypertension, stemming chiefly from the breakdown of proper tissue structure and function and aberrant cell migration during regenerative wound healing. A better understanding of these (impaired) PCP mechanisms is needed to fully harness the therapeutic opportunities of targeting PCP in chronic lung diseases.


Asunto(s)
Polaridad Celular , Enfermedades Pulmonares/metabolismo , Pulmón/crecimiento & desarrollo , Vía de Señalización Wnt , Animales , Movimiento Celular , Cilios/metabolismo , Citoesqueleto/metabolismo , Drosophila/metabolismo , Receptores Frizzled/metabolismo , Humanos , Ratones , Mucosa Respiratoria/metabolismo , Proteínas Wnt/metabolismo
20.
Am J Respir Crit Care Med ; 201(10): 1249-1262, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32023086

RESUMEN

Rationale: Bronchopulmonary dysplasia (BPD) is a leading complication of preterm birth that affects infants born in the saccular stage of lung development at <32 weeks of gestation. Although the mechanisms driving BPD remain uncertain, exposure to hyperoxia is thought to contribute to disease pathogenesis.Objectives: To determine the effects of hyperoxia on epithelial-mesenchymal interactions and to define the mediators of activated Wnt/ß-catenin signaling after hyperoxia injury.Methods: Three hyperoxia models were used: A three-dimensional organotypic coculture using primary human lung cells, precision-cut lung slices (PCLS), and a murine in vivo hyperoxia model. Comparisons of normoxia- and hyperoxia-exposed samples were made by real-time quantitative PCR, RNA in situ hybridization, quantitative confocal microscopy, and lung morphometry.Measurements and Main Results: Examination of an array of Wnt ligands in the three-dimensional organotypic coculture revealed increased mesenchymal expression of WNT5A. Inhibition of Wnt5A abrogated the BPD transcriptomic phenotype induced by hyperoxia. In the PCLS model, Wnt5A inhibition improved alveolarization following hyperoxia exposure, and treatment with recombinant Wnt5a reproduced features of the BPD phenotype in PCLS cultured in normoxic conditions. Chemical inhibition of NF-κB with BAY11-7082 reduced Wnt5a expression in the PCLS hyperoxia model and in vivo mouse hyperoxia model, with improved alveolarization in the PCLS model.Conclusions: Increased mesenchymal Wnt5A during saccular-stage hyperoxia injury contributes to the impaired alveolarization and septal thickening observed in BPD. Precise targeting of Wnt5A may represent a potential therapeutic strategy for the treatment of BPD.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Fibroblastos/metabolismo , Hiperoxia/genética , Pulmón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Wnt-5a/genética , Animales , Displasia Broncopulmonar , Técnicas de Cocultivo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Hiperoxia/metabolismo , Hibridación in Situ , Pulmón/crecimiento & desarrollo , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Microscopía Confocal , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Técnicas de Cultivo de Órganos , Reacción en Cadena en Tiempo Real de la Polimerasa , Sulfonas/farmacología , Proteína Wnt-5a/efectos de los fármacos , Proteína Wnt-5a/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA