Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.611
Filter
1.
Physiol Rep ; 12(14): e16143, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39034131

ABSTRACT

Inflammation through activation of caspase-1, seems to play a role in pulmonary hypertension induced by alveolar hypoxia. Whether alveolar hypoxia induces caspase-1-mediated inflammation and influx of leukocytes in other organs than the lungs, is not known. Our aim was to explore sites of caspase-1-related inflammation in alveolar hypoxia. Wild type (WT) mice were exposed to environmental hypoxia or room-air, and organs were analyzed. Right heart catheterization was performed after 14 days of alveolar hypoxia in WT mice and mice transplanted with WT or caspase-1-/- bone marrow. Hypoxia induced leukocyte accumulation and increased caspase-1 protein in the lungs, not in other organs. WT mice transplanted with WT or caspase-1-/- bone marrow showed no difference in pulmonary leukocyte accumulation or development of pulmonary hypertension after alveolar hypoxia. Caspase-1 and IL-18 were detected in bronchial epithelium in WT mice, and hypoxia induced IL-18 secretion from bronchial epithelial cells. IL-18 stimulation generated IL-6 mRNA in monocytes. Phosphorylated STAT3 was increased in hypoxic lungs, not in other organs. Alveolar hypoxia induces caspase-1 activation and leukocyte accumulation specific to the lungs, not in other organs. Caspase-1 activation and IL-18 secretion from bronchial epithelial cells might initiate hypoxia-induced inflammation, leading to pulmonary hypertension.


Subject(s)
Caspase 1 , Hypoxia , Inflammasomes , Interleukin-18 , Lung , Mice, Inbred C57BL , Animals , Male , Inflammasomes/metabolism , Mice , Caspase 1/metabolism , Caspase 1/genetics , Lung/metabolism , Lung/pathology , Interleukin-18/metabolism , Interleukin-18/genetics , Hypoxia/metabolism , Inflammation/metabolism , Inflammation/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Mice, Knockout , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology
2.
Nature ; 631(8021): 627-634, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987592

ABSTRACT

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


Subject(s)
Acute Lung Injury , Cell Lineage , Fibroblasts , Pneumonia , Pulmonary Alveoli , Pulmonary Fibrosis , Animals , Female , Humans , Male , Mice , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Cell Differentiation , Fibroblasts/pathology , Fibroblasts/metabolism , Homeostasis , Pneumonia/pathology , Pneumonia/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Stem Cell Niche , Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/pathology , Transforming Growth Factor beta/metabolism
3.
Biochem Biophys Res Commun ; 727: 150308, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38968769

ABSTRACT

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


Subject(s)
Autophagy , Glutamine , Lung Transplantation , Pulmonary Alveoli , Rats, Sprague-Dawley , Reperfusion Injury , Autophagy/drug effects , Animals , Glutamine/metabolism , Glutamine/pharmacology , Male , Humans , Mice , Rats , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Apoptosis/drug effects , Cell Line , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology
4.
Physiol Rep ; 12(11): e16057, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825580

ABSTRACT

The bronchoalveolar organoid (BAO) model is increasingly acknowledged as an ex-vivo platform that accurately emulates the structural and functional attributes of proximal airway tissue. The transition from bronchoalveolar progenitor cells to alveolar organoids is a common event during the generation of BAOs. However, there is a pressing need for comprehensive analysis to elucidate the molecular distinctions characterizing the pre-differentiated and post-differentiated states within BAO models. This study established a murine BAO model and subsequently triggered its differentiation. Thereafter, a suite of multidimensional analytical procedures was employed, including the morphological recognition and examination of organoids utilizing an established artificial intelligence (AI) image tracking system, quantification of cellular composition, proteomic profiling and immunoblots of selected proteins. Our investigation yielded a detailed evaluation of the morphologic, cellular, and molecular variances demarcating the pre- and post-differentiation phases of the BAO model. We also identified of a potential molecular signature reflective of the observed morphological transformations. The integration of cutting-edge AI-driven image analysis with traditional cellular and molecular investigative methods has illuminated key features of this nascent model.


Subject(s)
Cell Differentiation , Organoids , Organoids/metabolism , Organoids/cytology , Animals , Mice , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Artificial Intelligence , Proteomics/methods , Mice, Inbred C57BL
5.
Sci Adv ; 10(24): eado4791, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38865465

ABSTRACT

The stemness loss-associated dysregeneration of impaired alveolar type 2 epithelial (AT2) cells abolishes the reversible therapy of idiopathic pulmonary fibrosis (IPF). We here report an inhalable mucus-penetrating lipid nanoparticle (LNP) for codelivering dual mRNAs, promoting realveolarization via restoring AT2 stemness for IPF treatment. Inhalable LNPs were first formulated with dipalmitoylphosphatidylcholine and our in-house-made ionizable lipids for high-efficiency pulmonary mucus penetration and codelivery of dual messenger RNAs (mRNAs), encoding cytochrome b5 reductase 3 and bone morphogenetic protein 4, respectively. After being inhaled in a bleomycin model, LNPs reverses the mitochondrial dysfunction through ameliorating nicotinamide adenine dinucleotide biosynthesis, which inhibits the accelerated senescence of AT2 cells. Concurrently, pathological epithelial remodeling and fibroblast activation induced by impaired AT2 cells are terminated, ultimately prompting alveolar regeneration. Our data demonstrated that the mRNA-LNP system exhibited high protein expression in lung epithelial cells, which markedly extricated the alveolar collapse and prolonged the survival of fibrosis mice, providing a clinically viable strategy against IPF.


Subject(s)
Bleomycin , Mucus , Nanoparticles , Animals , Nanoparticles/chemistry , Mice , Mucus/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Disease Models, Animal , Administration, Inhalation , Lipids/chemistry , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , Liposomes
6.
J Pharmacol Toxicol Methods ; 128: 107526, 2024.
Article in English | MEDLINE | ID: mdl-38852686

ABSTRACT

INTRODUCTION: Inhalation of drugs for the treatment of pulmonary diseases has been used since a long time. Due to lungs' larger absorptive surface area, delivery of drugs to the lungs is the method of choice for different disorders. Here we present the establishment of a comprehensive permeability model using Type II alveolar epithelial cells and Beclomethasone Dipropionate (BDP) as a model drug delivered by pressurized metered dose inhaler (pMDI). METHODS: Using Type II alveolar epithelial cells, the method was standardized for parameters viz., cell density, viability, incubation period and membrane integrity. The delivery and deposition of drug were using the pMDI device with a Twin Stage Impinger (TSI) modified to accommodate cell culture insert having monolayer of cells. The analytical method for simultaneous estimation of BDP and Beclomathasone-17-Monopropionate (17-BMP) was validated as per the bioanalytical guidelines. The extent and rate of absorption of BDP was determined by quantifying the amount of drug permeated and the data represented by calculating its apparent permeability. RESULTS: Type II alveolar epithelial cells cultured at 0.55 × 105 cells/cm2 for 8-12 days under air-liquid interface were optimized for conducting permeability studies. The data obtained for absorptive transport showed a linear increase in the drug permeated against time for both BDP and 17-BMP along with proportional permeability profile. DISCUSSION: We have developed a robust in vitro model to study absorptive rate of drug transport across alveolar layer. Such models would create potential value during formulation development for comparative studies and selection of clinical candidates.


Subject(s)
Alveolar Epithelial Cells , Beclomethasone , Permeability , Administration, Inhalation , Beclomethasone/pharmacokinetics , Beclomethasone/administration & dosage , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Humans , Metered Dose Inhalers , Lung/metabolism , Lung/cytology , Lung/drug effects , Cells, Cultured , Cell Survival/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects
7.
Int J Numer Method Biomed Eng ; 40(8): e3839, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38885939

ABSTRACT

This study aims to investigate how inert gas affects the partial pressure of alveolar and venous blood using a fast and accurate operator splitting method (OSM). Unlike previous complex methods, such as the finite element method (FEM), OSM effectively separates governing equations into smaller sub-problems, facilitating a better understanding of inert gas transport and exchange between blood capillaries and surrounding tissue. The governing equations were discretized with a fully implicit finite difference method (FDM), which enables the use of larger time steps. The model employed partial differential equations, considering convection-diffusion in blood and only diffusion in tissue. The study explores the impact of initial arterial pressure, breathing frequency, blood flow velocity, solubility, and diffusivity on the partial pressure of inert gas in blood and tissue. Additionally, the effects of anesthetic inert gas and oxygen on venous blood partial pressure were analyzed. Simulation results demonstrate that the high solubility and diffusivity of anesthetic inert gas lead to its prolonged presence in blood and tissue, resulting in lower partial pressure in venous blood. These findings enhance our understanding of inert gas interaction with alveolar/venous blood, with potential implications for medical diagnostics and therapies.


Subject(s)
Noble Gases , Partial Pressure , Humans , Pulmonary Alveoli/physiology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/metabolism , Finite Element Analysis , Computer Simulation , Oxygen/blood , Oxygen/metabolism , Blood Flow Velocity/physiology , Diffusion
8.
Pflugers Arch ; 476(7): 1155-1168, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740599

ABSTRACT

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


Subject(s)
Melatonin , Mesenchymal Stem Cell Transplantation , Pulmonary Disease, Chronic Obstructive , Melatonin/pharmacology , Melatonin/administration & dosage , Animals , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Disease, Chronic Obstructive/metabolism , Mesenchymal Stem Cell Transplantation/methods , Rats , Male , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Physiologic/drug effects , Rats, Sprague-Dawley , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Lung/drug effects , Angiogenesis
9.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L160-L172, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38771132

ABSTRACT

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


Subject(s)
Alveolar Epithelial Cells , Chemokine CXCL10 , Receptors, CXCR3 , Animals , Mice , Alveolar Epithelial Cells/metabolism , Cell Proliferation , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Lung Injury/metabolism , Lung Injury/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Receptors, CXCR3/metabolism , Receptors, CXCR3/genetics , Regeneration , Signal Transduction
10.
Int J Biochem Cell Biol ; 172: 106587, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740281

ABSTRACT

Bronchopulmonary dysplasia (BPD) remains a significant challenge in neonatal care, the pathogenesis of which potentially involves altered lipid metabolism. Given the critical role of lipids in lung development and the injury response, we hypothesized that specific lipid species could serve as therapeutic agents in BPD. This study aimed to investigate the role of the lipid Phosphatidylcholine (PC) (16:0/14:0) in modulating BPD pathology and to elucidate its underlying mechanisms of action. Our approach integrated in vitro and in vivo methodologies to assess the effects of PC (16:0/14:0) on the histopathology, cellular proliferation, apoptosis, and molecular markers in lung tissue. In a hyperoxia-induced BPD rat model, we observed a reduction in alveolar number and an enlargement in alveolar size, which were ameliorated by PC (16:0/14:0) treatment. Correspondingly, in BPD cell models, PC (16:0/14:0) intervention led to increased cell viability, enhanced proliferation, reduced apoptosis, and elevated surfactant protein C (SPC) expression. RNA sequencing revealed significant gene expression differences between BPD and PC (16:0/14:0) treated groups, with a particular focus on Cldn1 (encoding claudin 1), which was significantly enriched in our analysis. Our findings suggest that PC (16:0/14:0) might protect against hyperoxia-induced alveolar type II cell damage by upregulating CLDN1 expression, potentially serving as a novel therapeutic target for BPD. This study not only advances our understanding of the role of lipids in BPD pathogenesis, but also highlights the significance of PC (16:0/14:0) in the prevention and treatment of BPD, offering new avenues for future research and therapeutic development.


Subject(s)
Alveolar Epithelial Cells , Bronchopulmonary Dysplasia , Claudin-1 , Hyperoxia , Phosphatidylcholines , Up-Regulation , Animals , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/etiology , Hyperoxia/metabolism , Hyperoxia/complications , Hyperoxia/pathology , Rats , Claudin-1/metabolism , Claudin-1/genetics , Phosphatidylcholines/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Rats, Sprague-Dawley , Apoptosis , Cell Proliferation , Humans , Pulmonary Alveoli/pathology , Pulmonary Alveoli/metabolism , Animals, Newborn , Disease Models, Animal
11.
Vet Anaesth Analg ; 51(4): 391-398, 2024.
Article in English | MEDLINE | ID: mdl-38719760

ABSTRACT

OBJECTIVE: To investigate the effect of three different doses of oral pregabalin on minimum alveolar concentration of isoflurane (MACISO) in cats. STUDY DESIGN: Prospective, randomized, placebo-controlled, blinded, crossover trial. ANIMALS: A group of eight healthy adult cats aged 24-48 months. METHODS: Cats were randomly assigned to three oral doses of pregabalin (low dose: 2.5 mg kg-1, medium dose: 5 mg kg-1, high dose: 10 mg kg-1) or placebo 2 hours before MACISO determination, with the multiple treatments administered with a minimum 7 day washout period. Anesthesia was induced and maintained with isoflurane in oxygen until endotracheal intubation was achieved, and maintained with isoflurane with volume-controlled ventilation. MACISO was determined in triplicate using the bracketing technique and tail clamp method 120 minutes after pregabalin or placebo administration. Physiologic variables (including heart rate and blood pressure) recorded during MACISO determination were averaged and compared between the pregabalin and placebo treatments. One-way analysis of variance and the Friedman test were used to assess the difference for normally and non-normally distributed data, respectively. The Tukey test was used as a post hoc analysis. Values of p < 0.05 were considered significant. RESULTS: The MACISO with the medium- and high-dose pregabalin treatments were 1.33 ± 0.21% and 1.23 ± 0.17%, respectively. These were significantly lower than MACISO after placebo treatment (1.62 ± 0.13%; p = 0.014, p < 0.001, respectively), representing a decrease of 18 ± 9% and 24 ± 6%. The mean plasma pregabalin concentration was negatively correlated with MACISO values. Physiologic variables did not differ significantly between treatments. CONCLUSIONS AND CLINICAL RELEVANCE: Doses of 5 or 10 mg kg-1 pregabalin, administered orally 2 hours before determining MACISO, had a significant isoflurane-sparing effect in cats.


Subject(s)
Anesthetics, Inhalation , Cross-Over Studies , Isoflurane , Pregabalin , Pulmonary Alveoli , Animals , Cats , Female , Male , Administration, Oral , Analgesics/administration & dosage , Analgesics/pharmacology , Analgesics/pharmacokinetics , Anesthesia, Inhalation/veterinary , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacokinetics , Anesthetics, Inhalation/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Isoflurane/administration & dosage , Isoflurane/pharmacokinetics , Pregabalin/administration & dosage , Pregabalin/pharmacology , Pulmonary Alveoli/metabolism
13.
PLoS One ; 19(5): e0300751, 2024.
Article in English | MEDLINE | ID: mdl-38717999

ABSTRACT

Transcriptional response to changes in oxygen concentration is mainly controlled by hypoxia-inducible transcription factors (HIFs). Besides regulation of hypoxia-responsible gene expression, HIF-3α has recently been shown to be involved in lung development and in the metabolic process of fat tissue. However, the precise mechanism for such properties of HIF-3α is still largely unknown. To this end, we generated HIF3A gene-disrupted mice by means of genome editing technology to explore the pleiotropic role of HIF-3α in development and physiology. We obtained adult mice carrying homozygous HIF3A gene mutations with comparable body weight and height to wild-type mice. However, the number of litters and ratio of homozygous mutation carriers born from the mating between homozygous mutant mice was lower than expected due to sporadic deaths on postnatal day 1. HIF3A gene-disrupted mice exhibited abnormal configuration of the lung such as a reduced number of alveoli and thickened alveolar walls. Transcriptome analysis showed, as well as genes associated with lung development, an upregulation of stearoyl-Coenzyme A desaturase 1, a pivotal enzyme for fatty acid metabolism. Analysis of fatty acid composition in the lung employing gas chromatography indicated an elevation in palmitoleic acid and a reduction in oleic acid, suggesting an imbalance in distribution of fatty acid, a constituent of lung surfactant. Accordingly, administration of glucocorticoid injections during pregnancy resulted in a restoration of normal alveolar counts and a decrease in neonatal mortality. In conclusion, these observations provide novel insights into a pivotal role of HIF-3α in the preservation of critically important structure and function of alveoli beyond the regulation of hypoxia-mediated gene expression.


Subject(s)
Apoptosis Regulatory Proteins , Pulmonary Alveoli , Repressor Proteins , Animals , Female , Male , Mice , Animals, Newborn , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Fatty Acids/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
14.
Curr Top Dev Biol ; 159: 59-129, 2024.
Article in English | MEDLINE | ID: mdl-38729684

ABSTRACT

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.


Subject(s)
Pulmonary Alveoli , Animals , Humans , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Pulmonary Gas Exchange/physiology , Regeneration , Lung/cytology , Lung/metabolism , Lung Injury/pathology
15.
Part Fibre Toxicol ; 21(1): 25, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760786

ABSTRACT

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


Subject(s)
Air Pollution, Indoor , Cell Survival , Particulate Matter , Humans , Air Pollution, Indoor/adverse effects , Particulate Matter/toxicity , Cell Survival/drug effects , A549 Cells , Cytokines/metabolism , THP-1 Cells , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Air Pollutants/toxicity , Inflammation/chemically induced , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology
16.
Part Fibre Toxicol ; 21(1): 26, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778339

ABSTRACT

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


Subject(s)
Particulate Matter , Pulmonary Alveoli , Vehicle Emissions , Humans , Vehicle Emissions/toxicity , Vehicle Emissions/analysis , A549 Cells , Particulate Matter/toxicity , Particulate Matter/analysis , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Particle Size , Microscopy, Electron, Transmission , Polyethylene Terephthalates/chemistry , Polyethylene Terephthalates/toxicity , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Air Pollutants/toxicity , Air Pollutants/analysis
17.
Am J Respir Cell Mol Biol ; 71(1): 30-42, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38579159

ABSTRACT

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


Subject(s)
Cell Communication , Cellular Microenvironment , Fibroblasts , Lung Injury , Pulmonary Alveoli , Humans , Animals , Lung Injury/pathology , Lung Injury/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrosis , Macrophages/metabolism , Macrophages/pathology
18.
Cell ; 187(10): 2428-2445.e20, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38579712

ABSTRACT

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.


Subject(s)
Alveolar Epithelial Cells , Lung , Stem Cells , Animals , Mice , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/cytology , Cell Differentiation , Cell Lineage , Lung/cytology , Lung/metabolism , Lung/physiology , Lung Injury/pathology , Mice, Inbred C57BL , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Receptors, Notch/metabolism , Regeneration , Signal Transduction , Stem Cells/metabolism , Stem Cells/cytology
19.
FASEB J ; 38(8): e23612, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648494

ABSTRACT

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


Subject(s)
Alveolar Epithelial Cells , Lung Injury , Regeneration , Humans , Regeneration/physiology , Animals , Lung Injury/metabolism , Lung Injury/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Stem Cells/metabolism , Stem Cells/physiology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/metabolism , Signal Transduction , Cell Differentiation
20.
Int J Cardiol ; 407: 132041, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38643800

ABSTRACT

BACKGROUND: In chronic heart failure (HF), exercise-induced increase in pulmonary capillary pressure may cause an increase of pulmonary congestion, or the development of pulmonary oedema. We sought to assess in HF patients the exercise-induced intra-thoracic fluid movements, by measuring plasma brain natriuretic peptide (BNP), lung comets and lung diffusion for carbon monoxide (DLCO) and nitric oxide (DLNO), as markers of hemodynamic load changes, interstitial space and alveolar-capillary membrane fluids, respectively. METHODS AND RESULTS: Twenty-four reduced ejection fraction HF patients underwent BNP, lung comets and DLCO/DLNO measurements before, at peak and 1 h after the end of a maximal cardiopulmonary exercise test. BNP significantly increased at peak from 549 (328-841) to 691 (382-1207, p < 0.0001) pg/mL and almost completely returned to baseline value 1 h after exercise. Comets number increased at peak from 9.4 ± 8.2 to 24.3 ± 16.7, returning to baseline (9.7 ± 7.4) after 1 h (p < 0.0001). DLCO did not change significantly at peak (from 18.01 ± 4.72 to 18.22 ± 4.73 mL/min/mmHg), but was significantly reduced at 1 h (16.97 ± 4.26 mL/min/mmHg) compared to both baseline (p = 0.0211) and peak (p = 0.0174). DLNO showed a not significant trend toward lower values 1 h post-exercise. CONCLUSIONS: Moderate/severe HF patients have a 2-step intra-thoracic fluid movement with exercise: the first during active exercise, from the vascular space toward the interstitial space, as confirmed by comets increase, without any effect on diffusion, and the second, during recovery, toward the alveolar-capillary membrane, clearing the interstitial space but worsening gas diffusion.


Subject(s)
Exercise Test , Exercise , Heart Failure , Pulmonary Alveoli , Humans , Heart Failure/physiopathology , Heart Failure/diagnostic imaging , Male , Female , Middle Aged , Exercise/physiology , Aged , Pulmonary Alveoli/physiopathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/diagnostic imaging , Exercise Test/methods , Capillaries/diagnostic imaging , Capillaries/physiopathology , Natriuretic Peptide, Brain/blood , Lung/diagnostic imaging , Lung/physiopathology , Lung/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL