Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Am J Respir Cell Mol Biol ; 70(5): 339-350, 2024 May.
Article in English | MEDLINE | ID: mdl-38207121

ABSTRACT

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


Subject(s)
Alveolar Epithelial Cells , Humans , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/ultrastructure , Cell Differentiation , Transcriptome , Cells, Cultured , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/cytology , Tight Junctions/metabolism
2.
Int J Mol Sci ; 22(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299227

ABSTRACT

Our previous study showed that in adult mice, conditional Nedd4-2-deficiency in club and alveolar epithelial type II (AE2) cells results in impaired mucociliary clearance, accumulation of Muc5b and progressive, terminal pulmonary fibrosis within 16 weeks. In the present study, we investigated ultrastructural alterations of the alveolar epithelium in relation to interstitial remodeling in alveolar septa as a function of disease progression. Two, eight and twelve weeks after induction of Nedd4-2 knockout, lungs were fixed and subjected to design-based stereological investigation at the light and electron microscopic level. Quantitative data did not show any abnormalities until 8 weeks compared to controls. At 12 weeks, however, volume of septal wall tissue increased while volume of acinar airspace and alveolar surface area significantly decreased. Volume and surface area of alveolar epithelial type I cells were reduced, which could not be compensated by a corresponding increase of AE2 cells. The volume of collagen fibrils in septal walls increased and was linked with an increase in blood-gas barrier thickness. A high correlation between parameters reflecting interstitial remodeling and abnormal AE2 cell ultrastructure could be established. Taken together, abnormal regeneration of the alveolar epithelium is correlated with interstitial septal wall remodeling.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/ultrastructure , Nedd4 Ubiquitin Protein Ligases/metabolism , Airway Remodeling/physiology , Alveolar Epithelial Cells/physiology , Animals , Epithelial Cells/metabolism , Female , Fibrosis/metabolism , Fibrosis/pathology , Lung/pathology , Male , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/genetics , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Surfactants , Respiratory Mucosa/metabolism
3.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073107

ABSTRACT

Inhaled Aspergillus fumigatus spores can be internalized by alveolar type II cells. Cell lines stably expressing fluorescently labeled components of endocytic pathway enable investigations of intracellular organization during conidia internalization and measurement of the process kinetics. The goal of this report was to evaluate the methodological appliance of cell lines for studying fungal conidia internalization. We have generated A549 cell lines stably expressing fluorescently labeled actin (LifeAct-mRuby2) and late endosomal protein (LAMP1-NeonGreen) following an evaluation of cell-pathogen interactions in live and fixed cells. Our data show that the LAMP1-NeonGreen cell line can be used to visualize conidia co-localization with LAMP1 in live and fixed cells. However, caution is necessary when using LifeAct-mRuby2-cell lines as it may affect the conidia internalization dynamics.


Subject(s)
Alveolar Epithelial Cells , Aspergillosis/microbiology , Aspergillus fumigatus , Host-Pathogen Interactions , A549 Cells , Alveolar Epithelial Cells/microbiology , Alveolar Epithelial Cells/ultrastructure , Aspergillus fumigatus/pathogenicity , Aspergillus fumigatus/physiology , Aspergillus fumigatus/ultrastructure , Green Fluorescent Proteins/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Optical Imaging , Phagocytosis , Spores, Fungal/metabolism
4.
BMC Pulm Med ; 21(1): 58, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33588817

ABSTRACT

BACKGROUND: Hyperoxia downregulates the tight junction (TJ) proteins of the alveolar epithelium and leads to barrier dysfunction. Previous study has showed that STE20/SPS1-related proline/alanine-rich kinase (SPAK) interferes with the intestinal barrier function in mice. The aim of the present study is to explore the association between SPAK and barrier function in the alveolar epithelium after hyperoxic exposure. METHODS: Hyperoxic acute lung injury (HALI) was induced by exposing mice to > 99% oxygen for 64 h. The mice were randomly allotted into four groups comprising two control groups and two hyperoxic groups with and without SPAK knockout. Mouse alveolar MLE-12 cells were cultured in control and hyperoxic conditions with or without SPAK knockdown. Transepithelial electric resistance and transwell monolayer permeability were measured for each group. In-cell western assay was used to screen the possible mechanism of p-SPAK being induced by hyperoxia. RESULTS: Compared with the control group, SPAK knockout mice had a lower protein level in the bronchoalveolar lavage fluid in HALI, which was correlated with a lower extent of TJ disruption according to transmission electron microscopy. Hyperoxia down-regulated claudin-18 in the alveolar epithelium, which was alleviated in SPAK knockout mice. In MLE-12 cells, hyperoxia up-regulated phosphorylated-SPAK by reactive oxygen species (ROS), which was inhibited by indomethacin. Compared with the control group, SPAK knockdown MLE-12 cells had higher transepithelial electrical resistance and lower transwell monolayer permeability after hyperoxic exposure. The expression of claudin-18 was suppressed by hyperoxia, and down-regulation of SPAK restored the expression of claudin-18. The process of SPAK suppressing the expression of claudin-18 and impairing the barrier function was mediated by p38 mitogen-activated protein kinase (MAPK). CONCLUSIONS: Hyperoxia up-regulates the SPAK-p38 MAPK signal pathway by ROS, which disrupts the TJ of the alveolar epithelium by suppressing the expression of claudin-18. The down-regulation of SPAK attenuates this process and protects the alveolar epithelium against the barrier dysfunction induced by hyperoxia.


Subject(s)
Acute Lung Injury/metabolism , Alveolar Epithelial Cells/metabolism , Claudins/genetics , Hyperoxia/metabolism , Protein Serine-Threonine Kinases/genetics , Pulmonary Alveoli/metabolism , Tight Junctions/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Acute Lung Injury/pathology , Alveolar Epithelial Cells/ultrastructure , Animals , Bronchoalveolar Lavage Fluid/chemistry , Claudins/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Knockdown Techniques , Hyperoxia/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Permeability , Protein Serine-Threonine Kinases/metabolism , Pulmonary Alveoli/ultrastructure , Reactive Oxygen Species/metabolism , Signal Transduction , Tight Junctions/ultrastructure
5.
Toxicol In Vitro ; 70: 105012, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33049313

ABSTRACT

Alveolar type II (ATII) epithelial cells contain lamellar bodies (LBs) which synthesize and store lung surfactants. In animals, the inhibition or knockout of leucine-rich repeat kinase 2 (LRRK2) causes abnormal enlargement of LBs in ATII cells. This effect of LRRK2 inhibition in lung is largely accepted as being mediated directly through blocking of the kinase function; however, downstream consequences in the lung remain unknown. In this work we established an in vitro alveolar epithelial cell (AEC) model that recapitulates the in vivo phenotype of ATII cells and developed an assay to quantify changes in LB size in response to LRRK2 inhibitors. Culture of primary human AECs at the air-liquid interface on matrigel and collagen-coated transwell inserts in the presence of growth factors promoted the LB formation and apical microvilli and induced expression of LRRK2 and ATII cell markers. Treatment with a selective LRRK2 inhibitor resulted in pharmacological reduction of phospho-LRRK2 and a significant increase in LB size; effects previously reported in lungs of non-human primates treated with LRRK2 inhibitor. In summary, our human in vitro AEC model recapitulates the abnormal lung findings observed in LRRK2-perturbed animals and holds the potential for expanding current understanding of LRRK2 function in the lung.


Subject(s)
Alveolar Epithelial Cells/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Models, Biological , ATP-Binding Cassette Transporters/metabolism , Adenocarcinoma of Lung/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/ultrastructure , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lung Neoplasms/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism
6.
Sci Rep ; 10(1): 20486, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33235275

ABSTRACT

To overcome the scarcity of primary human alveolar epithelial cells for lung research, and the limitations of current cell lines to recapitulate the phenotype, functional and molecular characteristics of the healthy human alveolar epithelium, we have developed a new method to immortalise primary human alveolar epithelial lung cells using a non-viral vector to transfect the telomerase catalytic subunit (hTERT) and the simian virus 40 large-tumour antigen (SV40). Twelve strains of immortalised cells (ICs) were generated and characterised using molecular, immunochemical and morphological techniques. Cell proliferation and sensitivity to polystyrene nanoparticles (PS) were evaluated. ICs expressed caveolin-1, podoplanin and receptor for advanced glycation end-products (RAGE), and most cells were negative for alkaline phosphatase staining, indicating characteristics of AT1-like cells. However, most strains also contained some cells that expressed pro-surfactant protein C, classically described to be expressed only by AT2 cells. Thus, the ICs mimic the cellular heterogeneity in the human alveolar epithelium. These ICs can be passaged, replicate rapidly and remain confluent beyond 15 days. ICs showed differential sensitivity to positive and negatively charged PS nanoparticles, illustrating their potential value as an in vitro model to study respiratory bioreactivity. These novel ICs offer a unique resource to study human alveolar epithelial biology.


Subject(s)
Alveolar Epithelial Cells/metabolism , Genetic Vectors/metabolism , Alkaline Phosphatase/metabolism , Alveolar Epithelial Cells/ultrastructure , Cell Line, Transformed , Cell Proliferation , Cell Respiration , Cell Survival , Cells, Cultured , Humans , Hydrodynamics , Lipids/chemistry , Nanoparticles/ultrastructure , Particle Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Static Electricity , Transcription, Genetic , Transfection
7.
Int J Mol Med ; 46(6): 2126-2136, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33125104

ABSTRACT

Bronchopulmonary dysplasia (BPD), also known as chronic lung disease, is one of the most common respiratory diseases in premature new­born humans. Mitochondria are not only the main source of reactive oxygen species but are also critical for the maintenance of homeostasis and a wide range of biological activities, such as producing energy, buffering cytosolic calcium and regulating signal transduction. However, as a critical quality control method for mitochondria, little is known about the role of mitophagy in BPD. The present study assessed mitochondrial function in hyperoxia­exposed alveolar type II (AT­II) cells of rats during lung development. New­born Sprague­Dawley rats were divided into hyperoxia (85% oxygen) and control (21% oxygen) groups. Histopathological and morphological properties of the lung tissues were assessed at postnatal days 1, 3, 7 and 14. Ultrastructural mitochondrial alteration was observed using transmission electron microscopy and the expression of the mitophagy proteins putative kinase (PINK)1, Parkin and Nip3­like protein X (NIX) in the lung tissues was evaluated using western blotting. Immunofluorescence staining was used to determine the co­localisation of PINK1 and Parkin. Real­time analyses of extracellular acidification rate and oxygen consumption rate were performed using primary AT­II cells to evaluate metabolic changes. Mitochondria in hyperoxia­exposed rat AT­II cells began to show abnormal morphological and physiological features. These changes were accompanied by decreased mitochondrial membrane potential and increased expression levels of PINK1­Parkin and NIX. Increased binding between a mitochondria marker (cytochrome C oxidase subunit IV isoform I) and an autophagy marker (microtubule­associated protein­1 light chain­3B) was observed in primary AT­II cells and was accompanied by decreased mitochondrial metabolic capacity in model rats. Thus, mitophagy mediated by PINK1, Parkin and NIX in AT­II cells occurred in hyperoxia­exposed new­born rats. These findings suggested that the accumulation of dysfunctional mitochondria may be a key factor in the pathogenesis of BPD and result in attenuated alveolar development.


Subject(s)
Hyperoxia/metabolism , Membrane Proteins/metabolism , Mitophagy , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pulmonary Alveoli/pathology , Ubiquitin-Protein Ligases/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/ultrastructure , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/complications , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Female , Hyperoxia/complications , Hyperoxia/pathology , Membrane Potential, Mitochondrial , Mitochondria/pathology , Mitochondria/ultrastructure , Pulmonary Alveoli/metabolism , Rats, Sprague-Dawley
8.
BMJ Case Rep ; 13(10)2020 Oct 10.
Article in English | MEDLINE | ID: mdl-33040033

ABSTRACT

The term baby presented with respiratory distress with X-ray pictures consistent as hyaline membrane disease (HMD). Baby was ventilated and treated with surfactant. Because of the persistence of high ventilation needs with X-ray pictures consistent with HMD with a transient response to surfactant every time, the possibility of an inherited disorder of surfactant metabolism was kept. Whole-exome sequencing revealed a novel homozygous missense mutation in the gene for ATP binding cassette transporter protein A3. The baby died after 100 days of ventilation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Pulmonary Surfactants/metabolism , Respiratory Distress Syndrome, Newborn/genetics , ATP-Binding Cassette Transporters/metabolism , Alveolar Epithelial Cells/ultrastructure , Biopsy , Continuous Positive Airway Pressure , DNA Mutational Analysis , Fatal Outcome , Female , Homozygote , Humans , Infant , Infant, Newborn , Infant, Premature , Intubation, Intratracheal , Lung/diagnostic imaging , Lung/pathology , Microscopy, Electron , Mutation, Missense , Radiography , Respiratory Distress Syndrome, Newborn/diagnosis , Respiratory Distress Syndrome, Newborn/pathology , Respiratory Distress Syndrome, Newborn/therapy , Severity of Illness Index , Exome Sequencing
10.
Rev Esp Patol ; 53(3): 188-192, 2020.
Article in English | MEDLINE | ID: mdl-32650970

ABSTRACT

The new coronavirus SARS-CoV-2, first identified in Wuhan, China in December, 2019, can cause Severe Acute Respiratory Syndrome (SARS) with massive alveolar damage and progressive respiratory failure. We present the relevant autopsy findings of the first patient known to have died from COVID19 pneumonia in Spain, carried out on the 14th of February, 2020, in our hospital (Hospital Arnau de Vilanova-Lliria, Valencia). Histological examination revealed typical changes of diffuse alveolar damage (DAD) in both the exudative and proliferative phase of acute lung injury. Intra-alveolar multinucleated giant cells, smudge cells and vascular thrombosis were present. The diagnosis was confirmed by reverse real-time PCR assay on a throat swab sample taken during the patient's admission. The positive result was reported fifteen days subsequent to autopsy.


Subject(s)
Autopsy , Betacoronavirus , Coronavirus Infections/pathology , Lung/pathology , Pandemics , Pneumonia, Viral/pathology , Respiratory Distress Syndrome/etiology , Aged , Alveolar Epithelial Cells/ultrastructure , Anion Exchange Protein 1, Erythrocyte/analysis , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Betacoronavirus/isolation & purification , COVID-19 , COVID-19 Testing , Carcinoma, Transitional Cell/complications , China , Clinical Laboratory Techniques , Community-Acquired Infections/diagnosis , Coronavirus Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , DNA-Binding Proteins/analysis , Humans , Lung/virology , Macrophages/chemistry , Macrophages/ultrastructure , Male , Pneumonia/diagnosis , Pneumonia, Viral/complications , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , Respiratory Distress Syndrome/pathology , SARS-CoV-2 , Spain/epidemiology , Transcription Factors/analysis , Travel , Urinary Bladder Neoplasms/complications
12.
Lancet ; 396(10247): 320-332, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32682491

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of an ongoing pandemic, with increasing deaths worldwide. To date, documentation of the histopathological features in fatal cases of the disease caused by SARS-CoV-2 (COVID-19) has been scarce due to sparse autopsy performance and incomplete organ sampling. We aimed to provide a clinicopathological report of severe COVID-19 cases by documenting histopathological changes and evidence of SARS-CoV-2 tissue tropism. METHODS: In this case series, patients with a positive antemortem or post-mortem SARS-CoV-2 result were considered eligible for enrolment. Post-mortem examinations were done on 14 people who died with COVID-19 at the King County Medical Examiner's Office (Seattle, WA, USA) and Snohomish County Medical Examiner's Office (Everett, WA, USA) in negative-pressure isolation suites during February and March, 2020. Clinical and laboratory data were reviewed. Tissue examination was done by light microscopy, immunohistochemistry, electron microscopy, and quantitative RT-PCR. FINDINGS: The median age of our cohort was 73·5 years (range 42-84; IQR 67·5-77·25). All patients had clinically significant comorbidities, the most common being hypertension, chronic kidney disease, obstructive sleep apnoea, and metabolic disease including diabetes and obesity. The major pulmonary finding was diffuse alveolar damage in the acute or organising phases, with five patients showing focal pulmonary microthrombi. Coronavirus-like particles were detected in the respiratory system, kidney, and gastrointestinal tract. Lymphocytic myocarditis was observed in one patient with viral RNA detected in the tissue. INTERPRETATION: The primary pathology observed in our cohort was diffuse alveolar damage, with virus located in the pneumocytes and tracheal epithelium. Microthrombi, where observed, were scarce and endotheliitis was not identified. Although other non-pulmonary organs showed susceptibility to infection, their contribution to the pathogenesis of SARS-CoV-2 infection requires further examination. FUNDING: None.


Subject(s)
Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/ultrastructure , Alveolar Epithelial Cells/virology , Autopsy , Betacoronavirus , COVID-19 , Coronavirus Infections/epidemiology , Female , Gastrointestinal Tract/pathology , Gastrointestinal Tract/ultrastructure , Gastrointestinal Tract/virology , Heart/virology , Humans , Kidney/pathology , Kidney/ultrastructure , Kidney/virology , Liver/pathology , Liver/ultrastructure , Liver/virology , Male , Middle Aged , Myocardium/pathology , Myocardium/ultrastructure , Pandemics , Pneumonia, Viral/epidemiology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/ultrastructure , Respiratory Mucosa/pathology , Respiratory Mucosa/ultrastructure , Respiratory Mucosa/virology , SARS-CoV-2 , Spleen/pathology , Spleen/ultrastructure , Spleen/virology , Thrombosis/pathology , Trachea/pathology , Trachea/ultrastructure , Trachea/virology , Washington/epidemiology
13.
Methods Cell Biol ; 159: 115-141, 2020.
Article in English | MEDLINE | ID: mdl-32586440

ABSTRACT

The lung consists of branched structures that are anatomically, developmentally and functionally divided into airway and alveolar regions. Each region contributes to lung-specific functions involving a defense system and gas exchange, and their dysfunction can cause fatal lung diseases. In the alveolar region, the cuboidal alveolar type 2 (AT2) cells account for 90% of the alveolar epithelial cells and serve as the tissue stem cells secreting pulmonary surfactant, and flattened alveolar type I (AT1) cells cover most of the alveolar surface directly contributing to gas exchange adjacent to capillary vessels. It has been difficult to culture alveolar epithelial cells in vitro, as the lineage-specific features of those cells are rapidly lost in a conventional two-dimensional culture setting. The culture of alveolar organoids (AOs) is an emerging technique that can help maintain the features of alveolar epithelial cells in vitro, and their application to human disease modeling is eagerly awaited. We herein describe our method of generating and culturing alveolar epithelial cells and AOs derived from human induced pluripotent stem cells (iPSCs). iPSCs derived from lung disease patients, including those with rare genetic diseases, will make help elucidate the disease mechanism and hopefully identify therapeutic targets.


Subject(s)
Alveolar Epithelial Cells/cytology , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Alveolar Epithelial Cells/ultrastructure , Cell Differentiation , Cells, Cultured , Feeder Cells/cytology , Fetus/cytology , Fibroblasts/cytology , Fluorescent Antibody Technique , Humans
14.
Int J Mol Sci ; 21(9)2020 Apr 27.
Article in English | MEDLINE | ID: mdl-32349261

ABSTRACT

Gas exchange in the lung takes place via the air-blood barrier in the septal walls of alveoli. The tissue elements that oxygen molecules have to cross are the alveolar epithelium, the interstitium and the capillary endothelium. The epithelium that lines the alveolar surface is covered by a thin and continuous liquid lining layer. Pulmonary surfactant acts at this air-liquid interface. By virtue of its biophysical and immunomodulatory functions, surfactant keeps alveoli open, dry and clean. What needs to be added to this picture is the glycocalyx of the alveolar epithelium. Here, we briefly review what is known about this glycocalyx and how it can be visualized using electron microscopy. The application of colloidal thorium dioxide as a staining agent reveals differences in the staining pattern between type I and type II alveolar epithelial cells and shows close associations of the glycocalyx with intraalveolar surfactant subtypes such as tubular myelin. These morphological findings indicate that specific spatial interactions between components of the surfactant system and those of the alveolar epithelial glycocalyx exist which may contribute to the maintenance of alveolar homeostasis, in particular to alveolar micromechanics, to the functional integrity of the air-blood barrier, to the regulation of the thickness and viscosity of the alveolar lining layer, and to the defence against inhaled pathogens. Exploring the alveolar epithelial glycocalyx in conjunction with the surfactant system opens novel physiological perspectives of potential clinical relevance for future research.


Subject(s)
Alveolar Epithelial Cells/metabolism , Glycocalyx/metabolism , Pulmonary Surfactants/metabolism , Respiratory Mucosa/metabolism , Alveolar Epithelial Cells/ultrastructure , Animals , Glycocalyx/ultrastructure , Humans , Pulmonary Alveoli/physiology , Pulmonary Alveoli/ultrastructure , Respiratory Mucosa/ultrastructure
15.
J Cell Mol Med ; 24(11): 6178-6190, 2020 06.
Article in English | MEDLINE | ID: mdl-32307881

ABSTRACT

Mycoplasma gallisepticum (MG) can cause chronic respiratory disease (CRD) in chickens. While several studies have reported the inflammatory functions of microRNAs during MG infection, the mechanism by which exosomal miRNAs regulate MG-induced inflammation remains to be elucidated. The expression of exosome-microRNA derived from MG-infected chicken type II pneumocytes (CP-II) was screened, and the target genes and function of differentially expressed miRNAs (DEGs) were predicted. To verify the role of exosomal gga-miR-451, Western blot, ELISA and RT-qPCR were used in this study. The results showed that a total of 722 miRNAs were identified from the two exosomal small RNA (sRNA) libraries, and 30 miRNAs (9 up-regulated and 21 down-regulated) were significantly differentially expressed. The target miRNAs were significantly enriched in the treatment group, such as cell cycle, Toll-like receptor signalling pathway and MAPK signalling pathway. The results have also confirmed that gga-miR-451-absent exosomes derived from MG-infected CP-II cells increased inflammatory cytokine production in chicken fibroblast cells (DF-1), and wild-type CP-II cell-derived exosomes displayed protective effects. Collectively, our work suggests that exosomes from MG-infected CP-II cells alter the dynamics of the DF-1 cells, and may contribute to pathology of the MG infection via exosomal gga-miR-451 targeting YWHAZ involving in inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Exosomes/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Inflammation/genetics , MicroRNAs/genetics , 14-3-3 Proteins/metabolism , Alveolar Epithelial Cells/ultrastructure , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line , Chickens/genetics , Cluster Analysis , Cytokines/metabolism , Exosomes/metabolism , Exosomes/ultrastructure , Gene Expression Regulation , Gene Regulatory Networks , Inflammation Mediators/metabolism , MicroRNAs/metabolism , Molecular Sequence Annotation , Reproducibility of Results , Toll-Like Receptors/metabolism
16.
Int J Mol Sci ; 21(3)2020 Jan 27.
Article in English | MEDLINE | ID: mdl-32012801

ABSTRACT

Acute lung injury (ALI) is characterized by enhanced permeability of the air-blood barrier, pulmonary edema, and hypoxemia. MicroRNA-21 (miR-21) was shown to be involved in pulmonary remodeling and the pathology of ALI, and we hypothesized that miR-21 knock-out (KO) reduces injury and remodeling in ALI. ALI was induced in miR-21 KO and C57BL/6N (wildtype, WT) mice by an intranasal administration of 75 µg lipopolysaccharide (LPS) in saline (n = 10 per group). The control mice received saline alone (n = 7 per group). After 24 h, lung function was measured. The lungs were then excised for proteomics, cytokine, and stereological analysis to address inflammatory signaling and structural damage. LPS exposure induced ALI in both strains, however, only WT mice showed increased tissue resistance and septal thickening upon LPS treatment. Septal alterations due to LPS exposure in WT mice consisted of an increase in extracellular matrix (ECM), including collagen fibrils, elastic fibers, and amorphous ECM. Proteomics analysis revealed that the inflammatory response was dampened in miR-21 KO mice with reduced platelet and neutrophil activation compared with WT mice. The WT mice showed more functional and structural changes and inflammatory signaling in ALI than miR-21 KO mice, confirming the hypothesis that miR-21 KO reduces the development of pathological changes in ALI.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Airway Remodeling/genetics , MicroRNAs/genetics , Pulmonary Alveoli/metabolism , Signal Transduction , Acute Lung Injury/pathology , Acute Lung Injury/physiopathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/ultrastructure , Animals , Chromatography, Liquid , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Male , Mass Spectrometry , Mice , Mice, Knockout , Pulmonary Alveoli/pathology , Pulmonary Alveoli/ultrastructure , RAW 264.7 Cells , Respiratory Function Tests
17.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480246

ABSTRACT

High surface tension at the alveolar air-liquid interface is a typical feature of acute and chronic lung injury. However, the manner in which high surface tension contributes to lung injury is not well understood. This study investigated the relationship between abnormal alveolar micromechanics, alveolar epithelial injury, intra-alveolar fluid properties and remodeling in the conditional surfactant protein B (SP-B) knockout mouse model. Measurements of pulmonary mechanics, broncho-alveolar lavage fluid (BAL), and design-based stereology were performed as a function of time of SP-B deficiency. After one day of SP-B deficiency the volume of alveolar fluid V(alvfluid,par) as well as BAL protein and albumin levels were normal while the surface area of injured alveolar epithelium S(AEinjure,sep) was significantly increased. Alveoli and alveolar surface area could be recruited by increasing the air inflation pressure. Quasi-static pressure-volume loops were characterized by an increased hysteresis while the inspiratory capacity was reduced. After 3 days, an increase in V(alvfluid,par) as well as BAL protein and albumin levels were linked with a failure of both alveolar recruitment and airway pressure-dependent redistribution of alveolar fluid. Over time, V(alvfluid,par) increased exponentially with S(AEinjure,sep). In conclusion, high surface tension induces alveolar epithelial injury prior to edema formation. After passing a threshold, epithelial injury results in vascular leakage and exponential accumulation of alveolar fluid critically hampering alveolar recruitability.


Subject(s)
Alveolar Epithelial Cells/pathology , Bronchoalveolar Lavage Fluid/chemistry , Pulmonary Surfactant-Associated Protein B/deficiency , Acinar Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/ultrastructure , Animals , Biomechanical Phenomena , Doxycycline/pharmacology , Female , Lung/drug effects , Lung/physiopathology , Lung/ultrastructure , Mice, Knockout , Models, Biological , Pulmonary Surfactant-Associated Protein B/metabolism , Structure-Activity Relationship , Surface Tension
18.
Sci Rep ; 9(1): 11973, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427591

ABSTRACT

Exocytosis is the intracellular trafficking step where a secretory vesicle fuses with the plasma membrane to release vesicle content. Actin and microtubules both play a role in exocytosis; however, their interplay is not understood. Here we study the interaction of actin and microtubules during exocytosis in lung alveolar type II (ATII) cells that secrete surfactant from large secretory vesicles. Surfactant extrusion is facilitated by an actin coat that forms on the vesicle shortly after fusion pore opening. Actin coat compression allows hydrophobic surfactant to be released from the vesicle. We show that microtubules are localized close to actin coats and stay close to the coats during their compression. Inhibition of microtubule polymerization by colchicine and nocodazole affected the kinetics of actin coat formation and the extent of actin polymerisation on fused vesicles. In addition, microtubule and actin cross-linking protein IQGAP1 localized to fused secretory vesicles and IQGAP1 silencing influenced actin polymerisation after vesicle fusion. This study demonstrates that microtubules can influence actin coat formation and actin polymerization on secretory vesicles during exocytosis.


Subject(s)
Actins/metabolism , Exocytosis/physiology , Microtubules/metabolism , Actins/genetics , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/ultrastructure , Animals , Biomarkers , Cell Membrane/metabolism , Coated Vesicles/drug effects , Coated Vesicles/metabolism , Fluorescent Antibody Technique , Male , Membrane Fusion , Microtubules/genetics , Protein Binding , Protein Transport , Rats , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Tubulin Modulators/pharmacology
19.
Lab Invest ; 99(6): 853-865, 2019 06.
Article in English | MEDLINE | ID: mdl-30728465

ABSTRACT

Although mammalian target of rapamycin inhibitors (mTORi) are used to treat various malignancies, they frequently induce active alveolitis and dyslipidemia. Abnormal lipid metabolism affects alveolar surfactant function and results in pulmonary disorders; however, the pathophysiology of lung injury and its relationship with lipid metabolism remain unknown. We investigated the relationship between lipid metabolism and alveolar epithelial injury, focusing on peroxisome proliferator-activated receptor-γ (PPAR-γ) as a lipid stress-related factor in mTORi-induced lung injury. We clinicopathologically examined three patients with mTORi-induced lung injury. We constructed an mTORi injury mouse model using temsirolimus in mice (30 mg/kg/day), with the vehicle control and bleomycin injury groups. We also constructed a cultured alveolar epithelial cell injury model using temsirolimus (0-40 µM) in the mouse lung epithelial cell line MLE-12 and performed analysis with or without pioglitazone (PPAR-γ agonist) treatment. All three patients had dyslipidemia and lung lesions of hyperplastic pneumocytes with foamy and enlarged changes. In the mouse model, temsirolimus induced significantly higher levels of total cholesterol and free fatty acids in serum and higher levels of surfactant protein D in serum and BAL fluid with an increase in inflammatory cytokines in the lung compared to control. Temsirolimus also induced hyperplastic foamy pneumocytes with increased lipid-associated spots and larger round electron-lucent bodies compared to the control or bleomycin groups in microscopic analyses. Multiple lipid-associated spots within the cytoplasm were also induced by temsirolimus administration in MLE-12 cells. Temsirolimus downregulated PPAR-γ expression in mouse lung and MLE-12 cells but upregulated cleaved caspase-3 in MLE-12 cells. Pioglitazone blocked the upregulated cleaved caspase-3 expression in MLE-12 cells. The pathogenesis of mTORi-induced lung disease may be involved in alveolar epithelial injury, via lipid metabolic stress associated with downregulated PPAR-γ expression. Focusing on the relationship between lipid metabolic stress and alveolar epithelial injury represents a potentially novel approach to the study of pulmonary damage.


Subject(s)
Alveolar Epithelial Cells/metabolism , Antineoplastic Agents/adverse effects , Everolimus/adverse effects , Lipid Metabolism , Lung Injury/chemically induced , Aged , Alveolar Epithelial Cells/ultrastructure , Animals , Cell Line , Cytokines/metabolism , Female , Humans , Hyperlipidemias/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice, Inbred C57BL , Middle Aged , PPAR gamma/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors
20.
Lab Invest ; 99(6): 830-852, 2019 06.
Article in English | MEDLINE | ID: mdl-30700849

ABSTRACT

In human idiopathic pulmonary fibrosis (IPF), collapse of distal airspaces occurs in areas of the lung not (yet) remodeled. Mice lungs overexpressing transforming growth factor-ß1 (TGF-ß1) recapitulate this abnormality: surfactant dysfunction results in alveolar collapse preceding fibrosis and loss of alveolar epithelial type II (AE2) cells' apical membrane surface area. Here we examined whether surfactant dysfunction-related alveolar collapse due to TGF-ß1 overexpression is linked to septal wall remodeling and AE2 cell abnormalities. Three and 6 days after gene transfer of TGF-ß1, mice received either intratracheal surfactant (Surf-groups: Curosurf®, 100 mg/kg bodyweight) or 0.9% NaCl (Saline-groups). On days 7 (D7) and 14 (D14), lung mechanics were assessed followed by design-based stereology at light and electron microscopic level to quantify structures. Compared with Saline, Surf showed significantly improved tissue elastance, increased numbers of open alveoli, as well as reduced alveolar size heterogeneity on D7. Deterioration in lung mechanics was highly correlated to the loss of open alveoli. On D14, lung mechanics, number of open alveoli, and alveolar size heterogeneity remained significantly improved in the Surf-group. Volumes of extracellular matrix and collagen fibrils in septal walls were significantly reduced, whereas the apical membrane surface area of AE2 cells was increased in Surf compared with Saline. In remodeled tissue with collapsed alveoli, three-dimensional reconstruction of AE2 cells based on scanning electron microscopy array tomography revealed that AE2 cells were trapped without contact to airspaces in the TGF-ß1 mouse model. Similar observations were made in human IPF. Based on correlation analyses, the number of open alveoli and of alveolar size heterogeneity were highly linked with the loss of apical membrane surface area of AE2 cells and deposition of collagen fibrils in septal walls on D14. In conclusion, surfactant replacement therapy stabilizes alveoli and prevents extracellular matrix deposition in septal walls in the TGF-ß1 model.


Subject(s)
Alveolar Epithelial Cells/drug effects , Pulmonary Fibrosis/prevention & control , Pulmonary Surfactants/therapeutic use , Airway Remodeling , Alveolar Epithelial Cells/ultrastructure , Animals , Disease Models, Animal , Female , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology , Pulmonary Surfactants/pharmacology , Respiratory Mechanics , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...