Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.331
Filtrar
1.
Nat Commun ; 15(1): 8112, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39284798

RESUMEN

While the critical role of NKX2-1 and its transcriptional targets in lung morphogenesis and pulmonary epithelial cell differentiation is increasingly known, mechanisms by which chromatin accessibility alters the epigenetic landscape and how NKX2-1 interacts with other co-activators required for alveolar epithelial cell differentiation and function are not well understood. Combined deletion of the histone methyl transferases Prdm3 and Prdm16 in early lung endoderm causes perinatal lethality due to respiratory failure from loss of AT2 cells and the accumulation of partially differentiated AT1 cells. Combination of single-cell RNA-seq, bulk ATAC-seq, and CUT&RUN data demonstrate that PRDM3 and PRDM16 regulate chromatin accessibility at NKX2-1 transcriptional targets critical for perinatal AT2 cell differentiation and surfactant homeostasis. Lineage specific deletion of PRDM3/16 in AT2 cells leads to lineage infidelity, with PRDM3/16 null cells acquiring partial AT1 fate. Together, these data demonstrate that NKX2-1-dependent regulation of alveolar epithelial cell differentiation is mediated by epigenomic modulation via PRDM3/16.


Asunto(s)
Células Epiteliales Alveolares , Diferenciación Celular , Cromatina , Proteínas de Unión al ADN , Factor Nuclear Tiroideo 1 , Factores de Transcripción , Animales , Factor Nuclear Tiroideo 1/metabolismo , Factor Nuclear Tiroideo 1/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cromatina/metabolismo , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Ratones Noqueados , Pulmón/citología , Pulmón/metabolismo , Linaje de la Célula , Femenino
2.
Stem Cell Res Ther ; 15(1): 273, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39218985

RESUMEN

BACKGROUND: Chronic lung disease of prematurity, called bronchopulmonary dysplasia (BPD), lacks effective therapies, stressing the need for preclinical testing systems that reflect human pathology for identifying causal pathways and testing novel compounds. Alveolar organoids derived from human pluripotent stem cells (hPSC) are promising test platforms for studying distal airway diseases like BPD, but current protocols do not accurately replicate the distal niche environment of the native lung. Herein, we investigated the contributions of cellular constituents of the alveolus and fetal respiratory movements on hPSC-derived alveolar organoid formation. METHODS: Human PSCs were differentiated in 2D culture into lung progenitor cells (LPC) which were then further differentiated into alveolar organoids before and after removal of co-developing mesodermal cells. LPCs were also differentiated in Transwell® co-cultures with and without human fetal lung fibroblast. Forming organoids were subjected to phasic mechanical strain using a Flexcell® system. Differentiation within organoids and Transwell® cultures was assessed by flow cytometry, immunofluorescence, and qPCR for lung epithelial and alveolar markers of differentiation including GATA binding protein 6 (GATA 6), E-cadherin (CDH1), NK2 Homeobox 1 (NKX2-1), HT2-280, surfactant proteins B (SFTPB) and C (SFTPC). RESULTS: We observed that co-developing mesenchymal progenitors promote alveolar epithelial type 2 cell (AEC2) differentiation within hPSC-derived lung organoids. This mesenchymal effect on AEC2 differentiation was corroborated by co-culturing hPSC-NKX2-1+ lung progenitors with human embryonic lung fibroblasts. The stimulatory effect did not require direct contact between fibroblasts and NKX2-1+ lung progenitors. Additionally, we demonstrate that episodic mechanical deformation of hPSC-derived lung organoids, mimicking in situ fetal respiratory movements, increased AEC2 differentiation without affecting proximal epithelial differentiation. CONCLUSION: Our data suggest that biophysical and mesenchymal components promote AEC2 differentiation within hPSC-derived distal organoids in vitro.


Asunto(s)
Diferenciación Celular , Pulmón , Organoides , Humanos , Organoides/citología , Organoides/metabolismo , Pulmón/citología , Pulmón/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Técnicas de Cocultivo/métodos , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo
3.
Physiol Rep ; 12(17): e70025, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39223101

RESUMEN

Major histocompatibility complex class I (MHC I) molecules present peptides to CD8+ T-cells for immunosurveillance of infection and cancer. Recent studies indicate lineage-specific heterogeneity in MHC I expression. While respiratory diseases rank among the leading causes of mortality, studies in mice have shown that lung epithelial cells (LECs) express the lowest levels of MHC I in the lung. This study aims to answer three questions: (i) Do human LECs express low levels of MHC I? (ii) Is LEC MHC I expression modulated in chronic respiratory diseases? (iii) Which factors regulate MHC I levels in human LECs? We analyzed human LECs from parenchymal explants using single-cell RNA sequencing and immunostaining. We confirmed low constitutive MHC I expression in human LECs, with significant upregulation in chronic respiratory diseases. We observed a sexual dimorphism, with males having higher MHC I levels under steady-state conditions, likely due to differential redox balance. Our study unveils the complex interplay between MHC I expression, sex, and respiratory disease. Since MHC I upregulation contributes to the development of immunopathologies in other models, we propose that it may have a similar impact on chronic lung disease.


Asunto(s)
Células Epiteliales , Antígenos de Histocompatibilidad Clase I , Pulmón , Humanos , Femenino , Masculino , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Pulmón/metabolismo , Pulmón/citología , Pulmón/inmunología , Células Epiteliales/metabolismo , Caracteres Sexuales , Enfermedades Pulmonares/metabolismo
4.
Molecules ; 29(18)2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39339380

RESUMEN

Cancer stem cells (CSCs) are most likely the main cause of lung cancer formation, metastasis, drug resistance, and genetic heterogeneity. Three-dimensional (3D) ex vivo cell culture models can facilitate stemness improvement and CSC enrichment. Considering the critical role of extracellular matrix (ECM) on CSC properties, the present study developed a thermo-responsive hydrogel using the porcine decellularized lung for 3D cell culture, and the cell-laden hydrogel culturing model was used to explore the CSC characteristics and potential utilization in CSC-specific drug evaluation. Results showed that the lung dECM hydrogel (LEH) was composed of the main ECM components and displayed excellent cellular compatibility. In addition, lung cancer cells 3D cultured in LEH displayed the overexpression of metastasis-related genes and enhanced migration properties, as compared with those in two-dimensional (2D) conditions. Notably, the CSC features, including the expression level of stemness-associated genes, colony formation capability, drug resistance, and the proportion of cancer stem-like cells (CD133+), were also enhanced in 3D cells. Furthermore, the attenuation effect of epigallocatechin gallate (EGCG) on CSC properties in the 3D model was observed, confirming the potential practicability of the 3D culture on CSC-targeted drug screening. Overall, our results suggest that the fabricated LEH is an effective and facile platform for 3D cell culture and CSC-specific drug evaluation.


Asunto(s)
Técnicas de Cultivo Tridimensional de Células , Hidrogeles , Neoplasias Pulmonares , Células Madre Neoplásicas , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Técnicas de Cultivo Tridimensional de Células/métodos , Animales , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Matriz Extracelular Descelularizada/farmacología , Matriz Extracelular Descelularizada/química , Pulmón/metabolismo , Pulmón/patología , Pulmón/citología , Porcinos , Catequina/análogos & derivados , Catequina/farmacología , Catequina/química , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos
5.
Sci Immunol ; 9(98): eadq7306, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39093959

RESUMEN

Short-lived repair-promoting macrophages are recruited to foci of lung damage during influenza infection-and they are Ly6G positive (see related Research Article by Ruscitti et al.).


Asunto(s)
Antígenos Ly , Macrófagos Alveolares , Animales , Antígenos Ly/metabolismo , Antígenos Ly/inmunología , Ratones , Macrófagos Alveolares/inmunología , Pulmón/inmunología , Pulmón/citología , Biomarcadores , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Humanos
6.
Nature ; 633(8029): 417-425, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39198650

RESUMEN

Severe defects in human IFNγ immunity predispose individuals to both Bacillus Calmette-Guérin disease and tuberculosis, whereas milder defects predispose only to tuberculosis1. Here we report two adults with recurrent pulmonary tuberculosis who are homozygous for a private loss-of-function TNF variant. Neither has any other clinical phenotype and both mount normal clinical and biological inflammatory responses. Their leukocytes, including monocytes and monocyte-derived macrophages (MDMs) do not produce TNF, even after stimulation with IFNγ. Blood leukocyte subset development is normal in these patients. However, an impairment in the respiratory burst was observed in granulocyte-macrophage colony-stimulating factor (GM-CSF)-matured MDMs and alveolar macrophage-like (AML) cells2 from both patients with TNF deficiency, TNF- or TNFR1-deficient induced pluripotent stem (iPS)-cell-derived GM-CSF-matured macrophages, and healthy control MDMs and AML cells differentiated with TNF blockers in vitro, and in lung macrophages treated with TNF blockers ex vivo. The stimulation of TNF-deficient iPS-cell-derived macrophages with TNF rescued the respiratory burst. These findings contrast with those for patients with inherited complete deficiency of the respiratory burst across all phagocytes, who are prone to multiple infections, including both Bacillus Calmette-Guérin disease and tuberculosis3. Human TNF is required for respiratory-burst-dependent immunity to Mycobacterium tuberculosis in macrophages but is surprisingly redundant otherwise, including for inflammation and immunity to weakly virulent mycobacteria and many other infectious agents.


Asunto(s)
Macrófagos , Tuberculosis Pulmonar , Factores de Necrosis Tumoral , Adulto , Femenino , Humanos , Masculino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Homocigoto , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/citología , Inflamación/inmunología , Interferón gamma/inmunología , Mutación con Pérdida de Función , Pulmón/citología , Pulmón/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Mycobacterium tuberculosis/inmunología , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Estallido Respiratorio , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/genética , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética , Adolescente , Adulto Joven
7.
Stem Cell Res Ther ; 15(1): 263, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39183355

RESUMEN

BACKGROUND: During pseudoglandular stage of the human lung development the primitive bronchial buds are initially conformed by simple tubules lined by endoderm-derived epithelium surrounded by mesenchyme, which will progressively branch into airways and start to form distal epithelial saculles. For first time alveolar type II (AT2) pneumocytes appears. This study aims to characterize the genes and microRNAs involved in this differentiation process and decipher its role in the starting alveolar differentiation. METHODS: Gene and microRNA profiling was performed in human embryonic lungs from 7 to 12 post conception weeks (pcw). Protein expression location of candidate genes were analyzed by immunofluorescense in embryonic lung tissue sections. mRNA/miRNA target pairs were identified using computational approaches and their expression was studied in purified epithelial/mesenchymal cell populations and in isolated tips and stalks from the bronchial tree. Additionally, silencing experiments in human embryonic lung mesenchymal cells and in human embryonic tip-derived lung organoids were performed, as well as organoid differentiation studies. AT2 cell markers were studied by qRT-PCR and by immunofluorescence. The TGFB-ß phosphorylated pathways was analyzed with membrane protein arrays. Lung explants were cultured in air/liquid interface with/without peptides. RESULTS: We identified 88 differentially expressed genes, including IGFBP3. Although IGFBP3 mRNA was detected in both epithelial and mesenchymal populations, the protein was restricted to the epithelium, indicating post-transcriptional regulation preventing IGFBP3 protein expression in the mesenchyme. MicroRNA profiling identified miR-34a as an IGFBP3 regulator. miR-34a was up-regulated in mesenchymal cells, and its silencing in human embryonic lung mesenchymal cells increased IGFBP3 levels. Additionally, IGFBP3 expression showed a marked downregulation from 7 to 12 pcw, suggesting its involvement in the differentiation process. The differentiation of human tip-derived lung embryonic organoids showed a drastic reduction in IGFBP3, supported by the scRNAseq data. IGFBP3 silencing in organoids activated an alveolar-like differentiation process characterized by stem cell markers downregulation and upregulation of AT2 markers. This process was mediated by TGFß signalling inhibition and BMP pathway activation. CONCLUSIONS: The IGFBP3/miR-34a axis restricts IGFBP3 expression in the embryonic undifferentiated lung epithelium, and the progressive downregulation of IGFBP3 during the pseudoglandular stage is required for alveolar differentiation.


Asunto(s)
Diferenciación Celular , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina , Pulmón , MicroARNs , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Pulmón/metabolismo , Pulmón/embriología , Pulmón/citología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología
8.
Chemosphere ; 364: 143205, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39214402

RESUMEN

The extensive use of synthetic antioxidants, notably N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine (6-PPD), in rubber-related products, particularly in tire manufacturing, has induced concerns regarding their environmental impact and potential health hazards. Despite the identification of 6-PPD and its derivative, 6-PPD quinone (6-PPDQ), in various water samples and their lethal effects on certain aquatic species (e.g., coho salmon, rainbow trout and brook trout), the levels of airborne 6-PPD/6-PPDQ and their respiratory toxicity remain relatively unexplored. In this study, we aimed to evaluate the respiratory toxicity potential of 6-PPD and its derivatives, with a specific focus on detecting these compounds in airborne particulates and assessing their toxic effects on lung cells. Characterization of four airborne fine particulate (FP) samples revealed spherical morphologies with diameters ranging from 17.7 to 225.7 nm, displaying slight agglomeration and negative surface charge. methanol/acetonitrile extraction followed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) analysis confirmed the presence of both 6-PPD and 6-PPDQ on the surfaces of FPs, with significant variations (0.26-1.05 µg g-1) in loading capacity observed among the samples. Subsequent exposure of lung cells (THP-1, BEAS-2B, and A549) to 6-PPD and 6-PPDQ revealed dose-dependent declines in mitochondrial metabolic activity induced by 6-PPD, along with severe membrane damage, ATP depletion, and pro-inflammatory cytokine release. Conversely, 6-PPDQ exhibited negligible toxicity in all tested parameters. These findings underscore the potential health risks associated with airborne 6-PPD exposure and emphasize the importance of further research into the respiratory toxicity of 6-PPD derivatives.


Asunto(s)
Pulmón , Material Particulado , Fenilendiaminas , Material Particulado/toxicidad , Material Particulado/análisis , Humanos , Pulmón/efectos de los fármacos , Pulmón/citología , Fenilendiaminas/toxicidad , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/análisis , Células A549 , Espectrometría de Masas en Tándem , Antioxidantes/metabolismo
9.
Am J Physiol Lung Cell Mol Physiol ; 327(4): L547-L556, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39189800

RESUMEN

Bronchial airways and lung parenchyma undergo both static and dynamic stretch in response to normal breathing as well as in the context of insults such as mechanical ventilation (MV) or in diseases such as asthma and chronic obstructive pulmonary disease (COPD) which lead to airway remodeling involving increased extracellular matrix (ECM) production. Here, the role of fibroblasts is critical, but the relationship between stretch- and fibroblast-induced ECM remodeling under these conditions is not well-explored. Piezo (PZ) channels play a role in mechanotransduction in many cell and organ systems, but their role in mechanical stretch-induced airway remodeling is not known. To explore this, we exposed human lung fibroblasts to 10% static stretch on a background of 5% oscillations for 48 h, with no static stretch considered controls. Collagen I, fibronectin, alpha-smooth muscle actin (α-SMA), and Piezo 1 (PZ1) expression was determined in the presence or absence of Yoda1 (PZ1 agonist) or GsMTx4 (PZ1 inhibitor). Collagen I, fibronectin, and α-SMA expression was increased by stretch and Yoda1, whereas pretreatment with GsMTx4 or knockdown of PZ1 by siRNA blunted this effect. Acute stretch in the presence and absence of Yoda1 demonstrated activation of the ERK pathway but not Smad. Measurement of [Ca2+]i responses to histamine showed significantly greater responses following stretch, effects that were blunted by knockdown of PZ1. Our findings identify an essential role for PZ1 in mechanical stretch-induced production of ECM mediated by ERK phosphorylation and Ca2+ influx in lung fibroblasts. Targeting PZ channels in fibroblasts may constitute a novel approach to ameliorate airway remodeling by decreasing ECM deposition.NEW & NOTEWORTHY The lung is an inherently mechanosensitive organ that can respond to mechanical forces in adaptive or maladaptive ways, including via remodeling resulting in increased fibrosis. We explored the mechanisms that link mechanical forces to remodeling using human lung fibroblasts. We found that mechanosensitive Piezo channels increase with stretch and mediate extracellular matrix formation and the fibroblast-to-myofibroblast transition that occurs with stretch. Our data highlight the importance of Piezo channels in lung mechanotransduction toward remodeling.


Asunto(s)
Fibroblastos , Canales Iónicos , Pulmón , Mecanotransducción Celular , Humanos , Pulmón/metabolismo , Pulmón/citología , Fibroblastos/metabolismo , Canales Iónicos/metabolismo , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Actinas/metabolismo , Células Cultivadas , Estrés Mecánico , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Calcio/metabolismo , Venenos de Araña , Péptidos y Proteínas de Señalización Intercelular
10.
STAR Protoc ; 5(3): 103183, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39093702

RESUMEN

Extracellular vesicles (EVs) are membranous nanoparticles classified based on their size and surface markers, which can be specific to various cell origins. Here, we present a protocol for the isolation of pulmonary-specific EVs in mice. We describe steps for differential centrifugation, density gradient centrifugation, and commercially available polyethylene glycol(PEG)-based precipitation, employing pulmonary-specific EV-bound chemicals and antibodies. We then detail procedures for the characterization of these EVs through nanoparticle tracking analysis, flow cytometry, scanning electron microscopy, and transmission electron microscopy. For complete details on the use and execution of this protocol, please refer to Lee et al.1,2,3,4.


Asunto(s)
Vesículas Extracelulares , Pulmón , Animales , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Ratones , Pulmón/citología , Pulmón/metabolismo , Citometría de Flujo/métodos , Microscopía Electrónica de Transmisión/métodos , Microscopía Electrónica de Rastreo/métodos , Centrifugación por Gradiente de Densidad/métodos , Nanopartículas/química , Polietilenglicoles/química
11.
Biotechnol Bioeng ; 121(9): 2848-2867, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39138873

RESUMEN

The fast-growing Chinese hamster lung (CHL)-YN cell line was recently developed for monoclonal antibody production. In this study, we applied a serum-free fed-batch cultivation process to immunoglobulin (Ig)G1-producing CHL-YN cells, which were then used to design a dynamic glucose supply system to stabilize the extracellular glucose concentration based on glucose consumption. Glucose consumption of the cultures rapidly oscillated following three phases of glutamine metabolism: consumption, production, and re-consumption. Use of the dynamic glucose supply prolonged the viability of the CHL-YN-IgG1 cell cultures and increased IgG1 production. Liquid chromatography with tandem mass spectrometry-based target metabolomics analysis of the extracellular metabolites during the first glutamine shift was conducted to search for depleted compounds. The results suggest that the levels of four amino acids, namely arginine, aspartate, methionine, and serine, were sharply decreased in CHL-YN cells during glutamine production. Supporting evidence from metabolic and gene expression analyses also suggest that CHL-YN cells acquired ornithine- and cystathionine-production abilities that differed from those in Chinese hamster ovary-K1 cells, potentially leading to proline and cysteine biosynthesis.


Asunto(s)
Anticuerpos Monoclonales , Cricetulus , Glucosa , Animales , Glucosa/metabolismo , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/metabolismo , Cricetinae , Línea Celular , Medio de Cultivo Libre de Suero , Metabolómica/métodos , Pulmón/metabolismo , Pulmón/citología , Metaboloma , Inmunoglobulina G/metabolismo , Células CHO , Técnicas de Cultivo Celular por Lotes/métodos , Glutamina/metabolismo
12.
J Vis Exp ; (209)2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39141526

RESUMEN

The identification and characterization of antigen-specific T cells during health and disease remains a key to improving our understanding of immune pathophysiology. The technical challenges of tracking antigen-specific T cell populations within the endogenous T cell repertoire have been greatly advanced by the development of peptide:MHC tetramer reagents. These fluorescently labeled soluble multimers of MHC class I or class II molecules complexed to antigenic peptide epitopes bind directly to T cells with corresponding T cell receptor (TCR) specificity and can, therefore, identify antigen-specific T cell populations in their native state without a requirement for a functional response induced by ex vivo stimulation. For exceedingly rare populations, tetramer-bound T cells can be magnetically enriched to increase the sensitivity and reliability of detection. As the investigation of tissue-resident T cell immunity deepens, there is a pressing need to identify antigen-specific T cells that traffic to and reside in nonlymphoid tissues. In this protocol, we present a detailed set of instructions for the isolation and characterization of antigen-specific T cells present within mouse lungs. This involves the isolation of T cells from digested lung tissue followed by a general T cell magnetic enrichment step and tetramer staining for flow cytometry analysis and sorting. The steps highlighted in this protocol utilize common techniques and readily available reagents, making it accessible for nearly any researcher engaged in mouse T cell immunology, and are highly adaptable for a variety of downstream analyses of any low frequency antigen-specific T cell population residing within the lungs.


Asunto(s)
Pulmón , Animales , Ratones , Pulmón/inmunología , Pulmón/citología , Péptidos/inmunología , Péptidos/química , Linfocitos T/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Epítopos de Linfocito T/inmunología
13.
Matrix Biol ; 132: 47-58, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39147560

RESUMEN

BACKGROUND: Lung fibroblasts play a central role in maintaining lung homeostasis and facilitating repair through the synthesis and organization of the extracellular matrix (ECM). This study investigated the cross-talk between interleukin-1 alpha (IL-1α) and transforming growth factor-ß (TGF-ß) signaling, two key regulators in tissue repair and fibrosis, in the context of lung fibroblast repair in the healthy lung. RESULTS: Stimulation of lung fibroblasts with TGF-ß1 and TGF-ß2 induced collagen-I and fibronectin protein expression (p < 0.05), a response inhibited with co-treatment with IL-1α (p < 0.05). Additionally, TGF-ß1 and TGF-ß2 induced myofibroblast differentiation, and collagen-I gel contraction, which were both suppressed by IL-1α (p < 0.05). In contrast, interleukin (IL)-6, IL-8 and thymic stromal lymphopoietin induced by IL-1α, were unaffected by TGF-ß1 or TGF-ß2. Mechanistically, IL-1α administration led to the suppression of TGF-ß1 and TGF-ß2 signaling, through downregulation of mRNA and protein for TGF-ß receptor II and the downstream adaptor protein TRAF6, but not through miR-146a that is known to be induced by IL-1α. DISCUSSION: IL-1α acts as a master regulator, modulating TGF-ß1 and TGF-ß2-induced ECM production, remodeling, and myofibroblast differentiation in human lung fibroblasts, playing a vital role in balancing tissue repair versus fibrosis. Further research is required to understand the dysregulated cross-talk between IL-1α and TGF-ß signaling in chronic lung diseases and the exploration of therapeutic opportunities. METHODS: Primary human lung fibroblasts (PHLF) were treated with media control, or 1 ng/ml IL-1α with or without 50 ng/ml TGF-ß1 or TGF-ß2 for 1, 6 and 72 h. Cell lysates were assessed for the expression of ECM proteins and signaling molecules by western blot, miRNA by qPCR, mRNA by RNA sequencing and cell supernatants for cytokine production by ELISA. PHLFs were also seeded in non-tethered collagen-I gels to measure contraction, and myofibroblast differentiation using confocal microscopy.


Asunto(s)
Matriz Extracelular , Fibroblastos , Interleucina-1alfa , Pulmón , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Humanos , Interleucina-1alfa/metabolismo , Interleucina-1alfa/genética , Matriz Extracelular/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Pulmón/metabolismo , Pulmón/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/citología , Diferenciación Celular , Miofibroblastos/metabolismo , Miofibroblastos/efectos de los fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , MicroARNs/genética , MicroARNs/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento Transformador beta2
14.
Luminescence ; 39(8): e4854, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39103184

RESUMEN

In this work, a benzofuranone-derived fluorescent probe BFSF was developed for imaging the sulphite level in living hypoxia pulmonary cells. Under the excitation of 510 nm, BFSF showed a strong fluorescence response at 570 nm when reacted with sulphite. In the solution system, the constructed hypercapnia and serious hypercapnia conditions did not affect the fluorescence response. In comparison with the recently reported probes, BFSF suggested the advantages including rapid response, steady signal reporting, high specificity and low cytotoxicity upon living lung cells. Under a normal incubation atmosphere, BFSF realized the imaging of both exogenous and endogenous sulphite in living pulmonary cells. In particular, BFSF achieved imaging the decrease of the sulphite level under severe hypoxia as well as the recovery of the sulphite level with urgent oxygen supplement. With the imaging capability for the sulphite level in living pulmonary cells under hypoxia conditions, BFSF together with the information herein was meaningful for investigating the anaesthesia-related biological indexes.


Asunto(s)
Benzofuranos , Colorantes Fluorescentes , Pulmón , Sulfitos , Colorantes Fluorescentes/química , Colorantes Fluorescentes/síntesis química , Benzofuranos/química , Benzofuranos/síntesis química , Sulfitos/análisis , Sulfitos/química , Pulmón/diagnóstico por imagen , Pulmón/citología , Humanos , Hipoxia de la Célula , Imagen Óptica , Estructura Molecular
15.
Life Sci ; 352: 122896, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38972632

RESUMEN

Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.


Asunto(s)
Carcinogénesis , Neoplasias Pulmonares , Macrófagos , Humanos , Animales , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Macrófagos/metabolismo , Pulmón/citología , Pulmón/fisiología , Macrófagos Alveolares/metabolismo , Progresión de la Enfermedad
16.
STAR Protoc ; 5(3): 103198, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39024094

RESUMEN

Mouse lung branching morphogenesis creates epithelial tree structures required for respiration. Here, we present a protocol for studying mouse lung developmental branching using lung explant cultures. We describe steps for isolating lungs with a video at embryonic day 12.5 (E12.5) and culturing as an explant for 2 days. We also detail procedures for microscopic imaging on days 0-2 and analysis of peripheral lung buds. This technique has the potential to investigate lung development in various conditions. For complete details on the use and execution of this protocol, please refer to Talvi et al.1.


Asunto(s)
Pulmón , Animales , Ratones , Pulmón/embriología , Pulmón/citología , Embrión de Mamíferos/citología , Técnicas de Cultivo de Órganos/métodos , Morfogénesis , Femenino
17.
STAR Protoc ; 5(3): 103201, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39028617

RESUMEN

Molecular and cellular mechanisms of human lung alveolar development are poorly understood due to a lack of in vitro model systems. This protocol details the isolation, derivation, and genetic modification of lung tip epithelial progenitors from human fetal lungs. It includes steps for isolating distal lung epithelial cells, expanding tip progenitor organoids, culturing tip organoids in vitro, and differentiating them into alveolar type 2 cells. This will aid in understanding alveolar differentiation mechanisms and neonatal diseases. For complete details on the use and execution of this protocol, please refer to Lim et al.1.


Asunto(s)
Diferenciación Celular , Pulmón , Organoides , Humanos , Diferenciación Celular/fisiología , Pulmón/citología , Pulmón/embriología , Organoides/citología , Organoides/metabolismo , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Células Madre/citología , Técnicas de Cultivo de Célula/métodos , Alveolos Pulmonares/citología , Alveolos Pulmonares/embriología , Células Cultivadas
18.
Physiol Rep ; 12(13): e16148, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38991987

RESUMEN

Pulmonary fibrosis is characterized by pathological accumulation of scar tissue in the lung parenchyma. Many of the processes that are implicated in fibrosis, including increased extracellular matrix synthesis, also occur following pneumonectomy (PNX), but PNX instead results in regenerative compensatory growth of the lung. As fibroblasts are the major cell type responsible for extracellular matrix production, we hypothesized that comparing fibroblast responses to PNX and bleomycin (BLM) would unveil key differences in the role they play during regenerative versus fibrotic lung responses. RNA-sequencing was performed on flow-sorted fibroblasts freshly isolated from mouse lungs 14 days after BLM, PNX, or sham controls. RNA-sequencing analysis revealed highly similar biological processes to be involved in fibroblast responses to both BLM and PNX, including TGF-ß1 and TNF-α. Interestingly, we observed smaller changes in gene expression after PNX than BLM at Day 14, suggesting that the fibroblast response to PNX may be muted by expression of transcripts that moderate pro-fibrotic pathways. Itpkc, encoding inositol triphosphate kinase C, was a gene uniquely up-regulated by PNX and not BLM. ITPKC overexpression in lung fibroblasts antagonized the pro-fibrotic effect of TGF-ß1. RNA-sequencing analysis has identified considerable overlap in transcriptional changes between fibroblasts following PNX and those overexpressing ITPKC.


Asunto(s)
Bleomicina , Fibroblastos , Ratones Endogámicos C57BL , Neumonectomía , Fibrosis Pulmonar , Bleomicina/farmacología , Animales , Fibroblastos/metabolismo , Fibroblastos/efectos de los fármacos , Ratones , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Pulmón/metabolismo , Pulmón/citología , Pulmón/patología , Masculino , Análisis de Secuencia de ARN/métodos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Células Cultivadas
19.
Aging (Albany NY) ; 16(13): 10694-10723, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38976646

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an age-related disease with poor prognosis and limited therapeutic options. Activation of lung fibroblasts and differentiation to myofibroblasts are the principal effectors of disease pathology, but damage and senescence of alveolar epithelial cells, specifically type II (ATII) cells, has recently been identified as a potential trigger event for the progressive disease cycle. Targeting ATII senescence and the senescence-associated secretory phenotype (SASP) is an attractive therapeutic strategy; however, translatable primary human cell models that enable mechanistic studies and drug development are lacking. Here, we describe a novel system of conditioned medium (CM) transfer from bleomycin-induced senescent primary alveolar epithelial cells (AEC) onto normal human lung fibroblasts (NHLF) that demonstrates an enhanced fibrotic transcriptional and secretory phenotype compared to non-senescent AEC CM treatment or direct bleomycin damage of the NHLFs. In this system, the bleomycin-treated AECs exhibit classical hallmarks of cellular senescence, including SASP and a gene expression profile that resembles aberrant epithelial cells of the IPF lung. Fibroblast activation by CM transfer is attenuated by pre-treatment of senescent AECs with the senolytic Navitoclax and AD80, but not with the standard of care agent Nintedanib or senomorphic JAK-targeting drugs (e.g., ABT-317, ruxolitinib). This model provides a relevant human system for profiling novel senescence-targeting therapeutics for IPF drug development.


Asunto(s)
Células Epiteliales Alveolares , Bleomicina , Senescencia Celular , Fibroblastos , Fibrosis Pulmonar Idiopática , Humanos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Bleomicina/toxicidad , Bleomicina/farmacología , Senescencia Celular/efectos de los fármacos , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/metabolismo , Medios de Cultivo Condicionados/farmacología , Indoles/farmacología , Fenotipo Secretor Asociado a la Senescencia/efectos de los fármacos , Pulmón/patología , Pulmón/citología , Pulmón/efectos de los fármacos , Sulfonamidas/farmacología , Senoterapéuticos/farmacología , Células Cultivadas , Pirimidinas/farmacología , Pirazoles/farmacología , Nitrilos/farmacología , Compuestos de Anilina
20.
Nat Commun ; 15(1): 5898, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003323

RESUMEN

Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.


Asunto(s)
Diferenciación Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística , Células Epiteliales , Feto , Pulmón , Humanos , Pulmón/embriología , Pulmón/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Feto/citología , Feto/embriología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Plasticidad de la Célula , Linaje de la Célula , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Análisis de la Célula Individual , Transcriptoma , Femenino , Regulación del Desarrollo de la Expresión Génica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA