Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 12.289
Filtrer
1.
Theranostics ; 14(9): 3603-3622, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948058

RÉSUMÉ

Background: Myofibroblasts (MYFs) are generally considered the principal culprits in excessive extracellular matrix deposition and scar formation in the pathogenesis of lung fibrosis. Lipofibroblasts (LIFs), on the other hand, are defined by their lipid-storing capacity and are predominantly found in the alveolar regions of the lung. They have been proposed to play a protective role in lung fibrosis. We previously reported that a LIF to MYF reversible differentiation switch occurred during fibrosis formation and resolution. In this study, we tested whether WI-38 cells, a human embryonic lung fibroblast cell line, could be used to study fibroblast differentiation towards the LIF or MYF phenotype and whether this could be relevant for idiopathic pulmonary fibrosis (IPF). Methods: Using WI-38 cells, Fibroblast (FIB) to MYF differentiation was triggered using TGF-ß1 treatment and FIB to LIF differentiation using Metformin treatment. We also analyzed the MYF to LIF and LIF to MYF differentiation by pre-treating the WI-38 cells with TGF-ß1 or Metformin respectively. We used IF, qPCR and bulk RNA-Seq to analyze the phenotypic and transcriptomic changes in the cells. We correlated our in vitro transcriptome data from WI-38 cells (obtained via bulk RNA sequencing) with the transcriptomic signature of LIFs and MYFs derived from the IPF cell atlas as well as with our own single-cell transcriptomic data from IPF patients-derived lung fibroblasts (LF-IPF) cultured in vitro. We also carried out alveolosphere assays to evaluate the ability of the proposed LIF and MYF cells to support the growth of alveolar epithelial type 2 cells. Results: WI-38 cells and LF-IPF display similar phenotypical and gene expression responses to TGF-ß1 and Metformin treatment. Bulk RNA-Seq analysis of WI-38 cells and LF-IPF treated with TGF-ß1, or Metformin indicate similar transcriptomic changes. We also show the partial conservation of the LIF and MYF signature extracted from the Habermann et al. scRNA-seq dataset in WI-38 cells treated with Metformin or TGF-ß1, respectively. Alveolosphere assays indicate that LIFs enhance organoid growth, while MYFs inhibit organoid growth. Finally, we provide evidence supporting the MYF to LIF and LIF to MYF reversible switch using WI-38 cells. Conclusions: WI-38 cells represent a versatile and reliable model to study the intricate dynamics of fibroblast differentiation towards the MYF or LIF phenotype associated with lung fibrosis formation and resolution, providing valuable insights to drive future research.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Fibrose pulmonaire idiopathique , Myofibroblastes , Facteur de croissance transformant bêta-1 , Humains , Myofibroblastes/métabolisme , Fibroblastes/métabolisme , Lignée cellulaire , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Poumon/anatomopathologie , Poumon/cytologie , Transcriptome , Metformine/pharmacologie , Plasticité cellulaire/effets des médicaments et des substances chimiques , Phénotype
2.
PLoS One ; 19(6): e0300772, 2024.
Article de Anglais | MEDLINE | ID: mdl-38913629

RÉSUMÉ

Gaseous and semi-volatile organic compounds emitted by the transport sector contribute to air pollution and have adverse effects on human health. To reduce harmful effects to the environment as well as to humans, renewable and sustainable bio-hybrid fuels are explored and investigated in the cluster of excellence "The Fuel Science Center" at RWTH Aachen University. However, data on the effects of bio-hybrid fuels on human health is scarce, leaving a data gap regarding their hazard potential. To help close this data gap, this study investigates potential toxic effects of a Ketone-Ester-Alcohol-Alkane (KEAA) fuel blend on A549 human lung cells. Experiments were performed using a commercially available air-liquid interface exposure system which was optimized beforehand. Then, cells were exposed at the air-liquid interface to 50-2000 ppm C3.7 of gaseous KEAA for 1 h. After a 24 h recovery period in the incubator, cells treated with 500 ppm C3.7 KEAA showed significant lower metabolic activity and cells treated with 50, 250, 500 and 1000 ppm C3.7 KEAA showed significant higher cytotoxicity compared to controls. Our data support the international occupational exposure limits of the single KEAA constituents. This finding applies only to the exposure scenario tested in this study and is difficult to extrapolate to the complex in vivo situation.


Sujet(s)
Poumon , Humains , Cellules A549 , Poumon/cytologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Biocarburants , Survie cellulaire/effets des médicaments et des substances chimiques , Gaz/toxicité , Composés organiques volatils/toxicité , Alcanes , Polluants atmosphériques/toxicité
3.
Eur J Pharmacol ; 977: 176711, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38839029

RÉSUMÉ

Histone deacetylase (HDAC) inhibitors are potential candidates for treating pulmonary fibrosis. MPT0E028, a novel pan-HDAC inhibitor, has been reported to exhibit antitumor activity in several cancer cell lines. In this study, we investigated the mechanism underlying the inhibitory effects of MPT0E028 on the expression of fibrogenic proteins in human lung fibroblasts (WI-38). Our results revealed that MPT0E028 inhibited transforming growth factor-ß (TGF-ß)-, thrombin-, and endothelin 1-induced connective tissue growth factor (CTGF) expression in a concentration-dependent manner. In addition, MPT0E028 suppressed TGF-ß-stimulated expression of fibronectin, collagen I, and α-smooth muscle actin (α-SMA). Furthermore, MPT0E028 inhibited the TGF-ß-induced phosphorylation of c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK). MPT0E028 reduced the increase in SMAD3 and c-Jun phosphorylation, and SMAD3-and activator protein-1 (AP-1)-luciferase activities under TGF-ß stimulation. Transfection with mitogen-activated protein kinase phosphatase-1 (MKP-1) siRNA reversed the suppressive effects of MPT0E028 on TGF-ß-induced increases in CTGF expression; JNK, p38, and ERK phosphorylation; and SMAD3 and AP-1 activation. Moreover, MPT0E028 increased MKP-1 acetylation and activity in WI-38 cells. Pretreatment with MPT0E028 reduced the fibrosis score and fibronectin, collagen, and α-SMA expression in bleomycin-induced pulmonary fibrosis mice. In conclusion, MPT0E028 induced MKP-1 acetylation and activation, which in turn inhibited TGF-ß-stimulated JNK, p38, and ERK phosphorylation; SMAD3 and AP-1 activation; and subsequent CTGF expression in human lung fibroblasts. Thus, MPT0E028 may be a potential drug for treating pulmonary fibrosis.


Sujet(s)
Facteur de croissance du tissu conjonctif , Dual Specificity Phosphatase 1 , Fibroblastes , Inhibiteurs de désacétylase d'histone , Poumon , Fibrose pulmonaire , Facteur de croissance transformant bêta , Facteur de croissance du tissu conjonctif/métabolisme , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/traitement médicamenteux , Animaux , Inhibiteurs de désacétylase d'histone/pharmacologie , Souris , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/cytologie , Poumon/métabolisme , Facteur de croissance transformant bêta/métabolisme , Dual Specificity Phosphatase 1/métabolisme , Dual Specificity Phosphatase 1/génétique , Lignée cellulaire , Protéine Smad-3/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Mâle , Activation enzymatique/effets des médicaments et des substances chimiques , Souris de lignée C57BL
4.
Methods Mol Biol ; 2813: 189-204, 2024.
Article de Anglais | MEDLINE | ID: mdl-38888779

RÉSUMÉ

Classic in vitro coculture assays of pathogens with host cells have contributed significantly to our understanding of the intracellular lifestyle of several pathogens. Coculture assays with pathogens and eukaryotic cells can be analyzed through various techniques including plating for colony-forming units (CFU), confocal microscopy, and flow cytometry. However, findings from in vitro assays require validation in an in vivo model. Several physiological conditions can influence host-pathogen interactions, which cannot easily be mimicked in vitro. Intravital microscopy (IVM) is emerging as a powerful tool for studying host-pathogen interactions by enabling in vivo imaging of living organisms. As a result, IVM has significantly enhanced the understanding of infection mediated by diverse pathogens. The versatility of IVM has also allowed for the imaging of various organs as sites of local infection. This chapter specifically focuses on IVM conducted on the lung for elucidating pulmonary immune response, primarily involving alveolar macrophages, to pathogens. Additionally, in this chapter we outline the protocol for lung IVM that utilizes a thoracic suction window to stabilize the lung for acquiring stable images.


Sujet(s)
Suivi cellulaire , Microscopie intravitale , Macrophages alvéolaires , Macrophages alvéolaires/cytologie , Microscopie intravitale/méthodes , Animaux , Suivi cellulaire/méthodes , Souris , Poumon/cytologie , Interactions hôte-pathogène
5.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 505-511, 2024 Jun 18.
Article de Chinois | MEDLINE | ID: mdl-38864137

RÉSUMÉ

OBJECTIVE: To investigate the effect of tofacitinib, a pan-Janus kinase (JAK) inhibitor, on transforming growth factor-beta 1 (TGF-ß1)-induced fibroblast to myofibroblast transition (FMT) and to explore its mechanism. To provide a theoretical basis for the clinical treatment of connective tissue disease-related interstitial lung disease (CTD-ILD). METHODS: (1) Human fetal lung fibroblast 1 (HFL-1) were cultured in vitro, and 6 groups were established: DMSO blank control group, TGF-ß1 induction group, and TGF-ß1 with different concentrations of tofacitinib (0.5, 1.0, 2.0, 5.0 µmol/L) drug intervention experimental groups. CCK-8 was used to measure the cell viability, and wound-healing assay was performed to measure cell migration ability. After 48 h of combined treatment, quantitative real-time PCR (RT-PCR) and Western blotting were used to detect the gene and protein expression levels of α-smooth muscle actin (α-SMA), fibronectin (FN), and collagen type Ⅰ (COL1). (2) RT-PCR and enzyme-linked immunosorbnent assay (ELISA) were used to detect the interleukin-6 (IL-6) gene and protein expression changes, respectively. (3) DMSO carrier controls, 1.0 µmol/L and 5.0 µmol/L tofacitinib were added to the cell culture media of different groups for pre-incubation for 30 min, and then TGF-ß1 was added to treat for 1 h, 6 h and 24 h. The phosphorylation levels of Smad2/3 and signal transducer and activator of transcription 3 (STAT3) protein were detected by Western blotting. RESULTS: (1) Tofacitinib inhibited the viability and migration ability of HFL-1 cells after TGF-ß1 induction. (2) The expression of α-SMA, COL1A1 and FN1 genes of HFL-1 in the TGF-ß1-induced groups was significantly up-regulated compared with the blank control group (P < 0.05). Compared with the TGF-ß1 induction group, α-SMA expression in the 5.0 µmol/L tofacitinib intervention group was significantly inhi-bited (P < 0.05). Compared with the TGF-ß1-induced group, FN1 gene was significantly inhibited in each intervention group at a concentration of 0.5-5.0 µmol/L (P < 0.05). Compared with the TGF-ß1-induced group, the COL1A1 gene expression in each intervention group did not change significantly. (3) Western blotting results showed that the protein levels of α-SMA and FN1 in the TGF-ß1-induced group were significantly higher than those in the control group (P < 0.05), and there was no significant difference in the expression of COL1A1. Compared with the TGF-ß1-induced group, the α-SMA protein level in the intervention groups with different concentrations decreased. And the differences between the TGF-ß1-induced group and 2.0 µmol/L or 5.0 µmol/L intervention groups were statistically significant (P < 0.05). Compared with the TGF-ß1-induced group, the FN1 protein levels in the intervention groups with different concentrations showed a downward trend, but the difference was not statistically significant. There was no difference in COL1A1 protein expression between the intervention groups compared with the TGF-ß1-induced group. (4) After TGF-ß1 acted on HFL-1 cells for 48 h, the gene expression of the IL-6 was up-regulated and IL-6 in culture supernatant was increased, the intervention with tofacitinib partly inhibited the TGF-ß1-induced IL-6 gene expression and IL-6 in culture supernatant. TGF-ß1 induced the increase of Smad2/3 protein phosphorylation in HFL-1 cells for 1 h and 6 h, STAT3 protein phosphorylation increased at 1 h, 6 h and 24 h, the pre-intervention with tofacitinib inhibited the TGF-ß1-induced Smad2/3 phosphorylation at 6 h and inhibited TGF-ß1-induced STAT3 phosphorylation at 1 h, 6 h and 24 h. CONCLUSION: Tofacitinib can inhibit the transformation of HFL-1 cells into myofibroblasts induced by TGF-ß1, and the mechanism may be through inhibiting the classic Smad2/3 pathway as well as the phosphorylation of STAT3 induced by TGF-ß1, thereby protecting the disease progression of pulmonary fibrosis.


Sujet(s)
Fibroblastes , Poumon , Myofibroblastes , Pipéridines , Pyrimidines , Facteur de transcription STAT-3 , Transduction du signal , Facteur de croissance transformant bêta-1 , Humains , Pyrimidines/pharmacologie , Pipéridines/pharmacologie , Facteur de transcription STAT-3/métabolisme , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Myofibroblastes/métabolisme , Myofibroblastes/cytologie , Myofibroblastes/effets des médicaments et des substances chimiques , Poumon/cytologie , Transduction du signal/effets des médicaments et des substances chimiques , Fibronectines/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Pyrroles/pharmacologie , Actines/métabolisme , Collagène de type I/métabolisme , Collagène de type I/génétique , Janus kinases/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Protéine Smad2/métabolisme , Pneumopathies interstitielles/métabolisme , Interleukine-6/métabolisme , Protéine Smad-3/métabolisme , Cellules cultivées
6.
Immunity ; 57(6): 1225-1242.e6, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38749446

RÉSUMÉ

Classical monocytes (CMs) are ephemeral myeloid immune cells that circulate in the blood. Emerging evidence suggests that CMs can have distinct ontogeny and originate from either granulocyte-monocyte- or monocyte-dendritic-cell progenitors (GMPs or MDPs). Here, we report surface markers that allowed segregation of murine GMP- and MDP-derived CMs, i.e., GMP-Mo and MDP-Mo, as well as their functional characterization, including fate definition following adoptive cell transfer. GMP-Mo and MDP-Mo yielded an equal increase in homeostatic CM progeny, such as blood-resident non-classical monocytes and gut macrophages; however, these cells differentially seeded various other selected tissues, including the dura mater and lung. Specifically, GMP-Mo and MDP-Mo differentiated into distinct interstitial lung macrophages, linking CM dichotomy to previously reported pulmonary macrophage heterogeneity. Collectively, we provide evidence for the existence of two functionally distinct CM subsets in the mouse that differentially contribute to peripheral tissue macrophage populations in homeostasis and following challenge.


Sujet(s)
Différenciation cellulaire , Macrophages , Monocytes , Animaux , Monocytes/immunologie , Monocytes/cytologie , Souris , Différenciation cellulaire/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Poumon/cytologie , Poumon/immunologie , Homéostasie , Souris de lignée C57BL , Cellules dendritiques/immunologie , Lignage cellulaire , Transfert adoptif
7.
Toxicol In Vitro ; 98: 105841, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38729454

RÉSUMÉ

3D cell culture models exposed at the air-liquid interface (ALI) represent a potential alternative to animal experiments for hazard and risk assessment of inhaled compounds. This study compares cocultures composed of either Calu-3, A549 or HBEC3-KT lung epithelial cells, cultured together with THP-1-derived macrophages and EA.hy926 endothelial cells, in terms of barrier capacity and responses to a standard reference sample of fine particulate matter (SRM 2786). High-content imaging analysis revealed a similar cellular composition between the different cell models. The 3D cell cultures with Calu-3 cells showed the greatest barrier capacity, as measured by transepithelial electrical resistance and permeability to Na-fluorescein. Mucus production was detected in 3D cell cultures based on Calu-3 and A549 cells. Exposure to SRM 2786 at ALI increased cytokine release and expression of genes associated with inflammation and xenobiotic metabolism. Moreover, the presence of THP-1-derived macrophages was central to the cytokine responses in all cell models. While the different 3D cell culture models produced qualitatively similar responses, more pronounced pro-inflammatory responses were observed in the basolateral compartment of the A549 and HBEC3-KT models compared to the Calu-3 model, likely due to their reduced barrier capacity and lower retention of secreted mediators in the apical compartment.


Sujet(s)
Cytokines , Poumon , Matière particulaire , Humains , Matière particulaire/toxicité , Poumon/effets des médicaments et des substances chimiques , Poumon/cytologie , Cytokines/métabolisme , Cytokines/génétique , Lignée cellulaire , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Techniques de culture cellulaire , Macrophages/effets des médicaments et des substances chimiques , Techniques de coculture , Polluants atmosphériques/toxicité , Mucus/métabolisme
8.
Curr Top Dev Biol ; 159: 59-129, 2024.
Article de Anglais | MEDLINE | ID: mdl-38729684

RÉSUMÉ

The mammalian lung completes its last step of development, alveologenesis, to generate sufficient surface area for gas exchange. In this process, multiple cell types that include alveolar epithelial cells, endothelial cells, and fibroblasts undergo coordinated cell proliferation, cell migration and/or contraction, cell shape changes, and cell-cell and cell-matrix interactions to produce the gas exchange unit: the alveolus. Full functioning of alveoli also involves immune cells and the lymphatic and autonomic nervous system. With the advent of lineage tracing, conditional gene inactivation, transcriptome analysis, live imaging, and lung organoids, our molecular understanding of alveologenesis has advanced significantly. In this review, we summarize the current knowledge of the constituents of the alveolus and the molecular pathways that control alveolar formation. We also discuss how insight into alveolar formation may inform us of alveolar repair/regeneration mechanisms following lung injury and the pathogenic processes that lead to loss of alveoli or tissue fibrosis.


Sujet(s)
Alvéoles pulmonaires , Animaux , Humains , Alvéoles pulmonaires/cytologie , Alvéoles pulmonaires/métabolisme , Échanges gazeux pulmonaires/physiologie , Régénération , Poumon/cytologie , Poumon/métabolisme , Lésion pulmonaire/anatomopathologie
9.
Cell Commun Signal ; 22(1): 289, 2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38802925

RÉSUMÉ

A cell is a dynamic system in which various processes occur simultaneously. In particular, intra- and intercellular signaling pathway crosstalk has a significant impact on a cell's life cycle, differentiation, proliferation, growth, regeneration, and, consequently, on the normal functioning of an entire organ. Hippo signaling and YAP/TAZ nucleocytoplasmic shuttling play a pivotal role in normal development, homeostasis, and tissue regeneration, particularly in lung cells. Intersignaling communication has a significant impact on the core components of the Hippo pathway and on YAP/TAZ localization. This review describes the crosstalk between Hippo signaling and key lung signaling pathways (WNT, SHH, TGFß, Notch, Rho, and mTOR) using lung cells as an example and highlights the remaining unanswered questions.


Sujet(s)
Poumon , Transduction du signal , Facteurs de transcription , Humains , Poumon/métabolisme , Poumon/cytologie , Animaux , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines de signalisation YAP/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Voie de signalisation Hippo , Espace intracellulaire/métabolisme
10.
Biosystems ; 240: 105216, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38692427

RÉSUMÉ

Cell shapes in tissues are affected by the biophysical interaction between cells. Tissue forces can influence specific cell features such as cell geometry and cell surface area. Here, we examined the 2-dimensional shape, size, and perimeter of pleural epithelial cells at various lung volumes. We demonstrated a 1.53-fold increase in 2-dimensional cell surface area and a 1.43-fold increase in cell perimeter at total lung capacity compared to residual lung volume. Consistent with previous results, close inspection of the pleura demonstrated wavy folds between pleural epithelial cells at all lung volumes. To investigate a potential explanation for the wavy folds, we developed a physical simulacrum suggested by D'Arcy Thompson in On Growth and Form. The simulacrum suggested that the wavy folds were the result of redundant cell membranes unable to contract. To test this hypothesis, we developed a numerical simulation to evaluate the impact of an increase in 2-dimensional cell surface area and cell perimeter on the shape of the cell-cell interface. Our simulation demonstrated that an increase in cell perimeter, rather than an increase in 2-dimensional cell surface area, had the most direct impact on the presence of wavy folds. We conclude that wavy folds between pleural epithelial cells reflects buckling forces arising from the excess cell perimeter necessary to accommodate visceral organ expansion.


Sujet(s)
Cellules épithéliales , Plèvre , Cellules épithéliales/physiologie , Cellules épithéliales/cytologie , Plèvre/cytologie , Plèvre/physiologie , Animaux , Forme de la cellule/physiologie , Humains , Poumon/cytologie , Poumon/physiologie , Modèles biologiques , Simulation numérique , Phénomènes biomécaniques/physiologie
11.
PLoS One ; 19(5): e0300902, 2024.
Article de Anglais | MEDLINE | ID: mdl-38748626

RÉSUMÉ

Tissue engineering predominantly relies on trial and error in vitro and ex vivo experiments to develop protocols and bioreactors to generate functional tissues. As an alternative, in silico methods have the potential to significantly reduce the timelines and costs of experimental programs for tissue engineering. In this paper, we propose a methodology to formulate, select, calibrate, and test mathematical models to predict cell population growth as a function of the biochemical environment and to design optimal experimental protocols for model inference of in silico model parameters. We systematically combine methods from the experimental design, mathematical statistics, and optimization literature to develop unique and explainable mathematical models for cell population dynamics. The proposed methodology is applied to the development of this first published model for a population of the airway-relevant bronchio-alveolar epithelial (BEAS-2B) cell line as a function of the concentration of metabolic-related biochemical substrates. The resulting model is a system of ordinary differential equations that predict the temporal dynamics of BEAS-2B cell populations as a function of the initial seeded cell population and the glucose, oxygen, and lactate concentrations in the growth media, using seven parameters rigorously inferred from optimally designed in vitro experiments.


Sujet(s)
Prolifération cellulaire , Simulation numérique , Poumon , Modèles biologiques , Humains , Lignée cellulaire , Poumon/cytologie , Poumon/métabolisme , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Ingénierie tissulaire/méthodes , Glucose/métabolisme , Oxygène/métabolisme
12.
Methods Mol Biol ; 2775: 385-391, 2024.
Article de Anglais | MEDLINE | ID: mdl-38758332

RÉSUMÉ

A special feature of the human fungal pathogen Cryptococcus neoformans is its morphological changes triggered by the interaction with the host. During infection, a specific increase in cell size is observed, particularly in lung tissue, from a typical cell size of 5-7 µm cells to cells larger than 10 µm, dubbed titan cells (TCs). However, the study of this specific cell subpopulation was, until now, only possible via recovery of TCs from lungs of mice during experimental infections where stable and reproducible generation of TCs occurs.The protocol described here generates TCs using in vitro conditions and measures cell size using a rapid, automated method. TC generation in vitro is robust and reproducible, generating yeast cells harboring the same characteristics of TCs generated in vivo.


Sujet(s)
Cryptococcus neoformans , Cryptococcus neoformans/cytologie , Cryptococcus neoformans/physiologie , Animaux , Souris , Cryptococcose/microbiologie , Taille de la cellule , Poumon/microbiologie , Poumon/cytologie , Humains
13.
Sci Immunol ; 9(95): eadj2654, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38820141

RÉSUMÉ

Tissue-resident innate lymphoid cells (ILCs) play a vital role in the frontline defense of various tissues, including the lung. The development of type 2 ILCs (ILC2s) depends on transcription factors such as GATA3, RORα, GFI1, and Bcl11b; however, the factors regulating lung-resident ILC2s remain unclear. Through fate mapping analysis of the paralog transcription factors GFI1 and GFI1B, we show that GFI1 is consistently expressed during the transition from progenitor to mature ILC2s. In contrast, GFI1B expression is limited to specific subsets of bone marrow progenitors and lung-resident ILC progenitors. We found that GFI1B+ lung ILC progenitors represent a multi-lineage subset with tissue-resident characteristics and the potential to form lung-derived ILC subsets and liver-resident ILC1s. Loss of GFI1B in bone marrow progenitors led to the selective loss of lung-resident IL-18R+ ILCs and mature ILC2, subsequently preventing the emergence of effector ILCs that could protect the lung against inflammatory or tumor challenge.


Sujet(s)
Immunité innée , Poumon , Souris de lignée C57BL , Protéines proto-oncogènes , Animaux , Poumon/immunologie , Poumon/cytologie , Souris , Immunité innée/immunologie , Protéines proto-oncogènes/immunologie , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/déficit , Protéines proto-oncogènes/métabolisme , Progéniteurs lymphoïdes/immunologie , Progéniteurs lymphoïdes/cytologie , Protéines de répression/génétique , Protéines de répression/immunologie , Souris knockout , Lymphocytes/immunologie , Différenciation cellulaire/immunologie , Protéines de liaison à l'ADN , Facteurs de transcription
14.
Commun Biol ; 7(1): 665, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38816547

RÉSUMÉ

The evolution and development of vertebrate lungs have been widely studied due to their significance in terrestrial adaptation. Amphibians possess the most primitive lungs among tetrapods, underscoring their evolutionary importance in bridging the transition from aquatic to terrestrial life. However, the intricate process of cell differentiation during amphibian lung development remains poorly understood. Using single-cell RNA sequencing, we identify 13 cell types in the developing lungs of a land-dwelling frog (Microhyla fissipes). We elucidate the differentiation trajectories and mechanisms of mesenchymal cells, identifying five cell fates and their respective driver genes. Using temporal dynamics analyses, we reveal the gene expression switches of epithelial cells, which facilitate air breathing during metamorphosis. Furthermore, by integrating the published data from another amphibian and two terrestrial mammals, we illuminate both conserved and divergent cellular repertoires during the evolution of tetrapod lungs. These findings uncover the frog lung cell differentiation trajectories and functionalization for breathing in air and provide valuable insights into the cell-type evolution of vertebrate lungs.


Sujet(s)
Anura , Différenciation cellulaire , Poumon , Analyse sur cellule unique , Animaux , Poumon/cytologie , Poumon/physiologie , Analyse sur cellule unique/méthodes , Anura/physiologie , Respiration , Métamorphose biologique , Régulation de l'expression des gènes au cours du développement , Analyse de séquence d'ARN/méthodes
15.
Development ; 151(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38738653

RÉSUMÉ

During alveologenesis, multiple mesenchymal cell types play crucial roles in maximising the lung surface area. In their study, David Ornitz and colleagues define the repertoire of lung fibroblasts, with a particular focus on alveolar myofibroblasts. To know more about their work, we spoke to the first author, Yongjun Yin, and the corresponding author, David Ornitz, Alumni Endowed Professor at the Department of Developmental Biology, Washington University School of Medicine, St. Louis.


Sujet(s)
Biologie du développement , Humains , Histoire du 21ème siècle , Biologie du développement/histoire , Histoire du 20ème siècle , Poumon/embryologie , Poumon/métabolisme , Poumon/cytologie , Animaux
16.
Talanta ; 274: 126066, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38599125

RÉSUMÉ

The evaluation of nanoparticles (NPs) cytotoxicity is crucial for advancing nanotechnology and assessing environmental pollution. However, existing methods for NPs cytotoxicity evaluation suffer from limited accuracy and inadequate information content. In the study, we developed a novel detection platform that enables the identification of cellular carbonyl metabolites at the organ level. The platform is integrated with a cell co-culture lung organ chip (LOC) and a micropillar concentrator. Notably, our work represents the successful measurement of the amounts of cellular metabolites on LOC system. The volatile carbonyl metabolites (VCMs) generated by cells exposure to various types of NPs with different concentrations were captured and detected by high-resolution mass spectrometry (MS). Compared with conventional cell viability and reactive oxygen species (ROS) analysis, our method discerns the toxicological impact of NPs at low concentrations by analyzed VCM at levels as low as ppb level. The LOC system based metabolic gas detection confirmed that low concentrations of NPs have a toxic effect on the cell model, which was not reflected in the fluorescence detection, and the effect of NP material is more significant than the size effect. Furthermore, this method can distinguish different NPs acting on cell models through cluster analysis of multiple VCMs.


Sujet(s)
Laboratoires sur puces , Poumon , Nanoparticules , Composés organiques volatils , Humains , Poumon/cytologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Composés organiques volatils/analyse , Composés organiques volatils/métabolisme , Nanoparticules/composition chimique , Nanoparticules/toxicité , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules A549 , Espèces réactives de l'oxygène/métabolisme , Espèces réactives de l'oxygène/analyse , Systèmes microphysiologiques
17.
Development ; 151(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38602479

RÉSUMÉ

Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFBs) and a stable but poorly described population of lipid-rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFBs). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single-cell RNA sequencing and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a MyoFB differentiation program that is distinct from other mesenchymal cell types and increases the known repertoire of mesenchymal cell types in the neonatal lung.


Sujet(s)
Animaux nouveau-nés , Différenciation cellulaire , Poumon , Myofibroblastes , Animaux , Myofibroblastes/métabolisme , Myofibroblastes/cytologie , Souris , Poumon/cytologie , Poumon/embryologie , Poumon/métabolisme , Lignage cellulaire , Organogenèse , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme
18.
Cell ; 187(10): 2428-2445.e20, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38579712

RÉSUMÉ

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.


Sujet(s)
Pneumocytes , Poumon , Cellules souches , Animaux , Souris , Pneumocytes/métabolisme , Pneumocytes/cytologie , Différenciation cellulaire , Lignage cellulaire , Poumon/cytologie , Poumon/métabolisme , Poumon/physiologie , Lésion pulmonaire/anatomopathologie , Souris de lignée C57BL , Alvéoles pulmonaires/cytologie , Alvéoles pulmonaires/métabolisme , Récepteurs Notch/métabolisme , Régénération , Transduction du signal , Cellules souches/métabolisme , Cellules souches/cytologie
19.
Toxicol In Vitro ; 98: 105828, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621549

RÉSUMÉ

Fine particulates in city air significantly impact human health, but the hazardous compositional mechanisms are still unclear. Besides the toxicity of environmental PM2.5 to in vitro human lung epithelial cells (A549), the independent cytotoxicity of PM2.5-bound water-soluble (WS-PM2.5) and water-insoluble (WIS-PM2.5) fractions were also compared by cell viability, oxidative stress (reactive oxygen species, ROS), and inflammatory injury (IL-6 and TNF-α). The cytotoxicity of PM2.5 varied significantly by sampling season and place, with degrees greater in winter and spring than in summer and autumn, related to corresponding trend of air PM2.5 level, and also higher in industrial than urban site, although their PM2.5 pollution levels were comparable. The PM2.5 bound metals (Ni, Cr, Fe, and Mn) may contribute to cellular injury. Both WS-PM2.5 and WIS-PM2.5 posed significant cytotoxicity, that WS-PM2.5 was more harmful than WIS-PM2.5 in terms of decreasing cell viability and increasing inflammatory cytokines production. In particular, industrial samples were usually more toxic than urban samples, and those from summer were generally less toxic than other seasons. Hence, in order to mitigate the health risks of PM2.5 pollution, the crucial targets might be components of heavy metals and soluble fractions, and sources in industrial areas, especially during the cold seasons.


Sujet(s)
Polluants atmosphériques , Survie cellulaire , Poumon , Matière particulaire , Espèces réactives de l'oxygène , Humains , Matière particulaire/toxicité , Survie cellulaire/effets des médicaments et des substances chimiques , Polluants atmosphériques/toxicité , Cellules A549 , Poumon/effets des médicaments et des substances chimiques , Poumon/cytologie , Espèces réactives de l'oxygène/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Solubilité , Interleukine-6/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Eau/composition chimique , Saisons
20.
Tissue Eng Regen Med ; 21(5): 653-671, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38466362

RÉSUMÉ

Organoids are essentially an in vitro (lab-grown) three-dimensional tissue culture system model that meticulously replicates the structure and physiology of human organs. A few of the present applications of organoids are in the basic biological research area, molecular medicine and pharmaceutical drug testing. Organoids are crucial in connecting the gap between animal models and human clinical trials during the drug discovery process, which significantly lowers the time duration and cost associated with each stage of testing. Likewise, they can be used to understand cell-to-cell interactions, a crucial aspect of tissue biology and regeneration, and to model disease pathogenesis at various stages of the disease. Lung organoids can be utilized to explore numerous pathophysiological activities of a lung since they share similarities with its function. Researchers have been trying to recreate the complex nature of the lung by developing various "Lung organoids" models.This article is a systematic review of various developments of lung organoids and their potential progenitors. It also covers the in-depth applications of lung organoids for the advancement of translational research. The review discusses the methodologies to establish different types of lung organoids for studying the regenerative capability of the respiratory system and comprehending various respiratory diseases.Respiratory diseases are among the most common worldwide, and the growing burden must be addressed instantaneously. Lung organoids along with diverse bio-engineering tools and technologies will serve as a novel model for studying the pathophysiology of various respiratory diseases and for drug screening purposes.


Sujet(s)
Poumon , Organoïdes , Organoïdes/cytologie , Humains , Poumon/cytologie , Animaux , Ingénierie tissulaire/méthodes , Régénération , Médecine régénérative/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...