451 results on '"Lung epithelium"'
Search Results
2. Microphysiological Models of Lung Epithelium‐Alveolar Macrophage Co‐Cultures to Study Chronic Lung Disease.
- Author
-
Lagowala, Dave A., Wally, Arabelis, Wilmsen, Kai, Kim, Byunggik, Yeung‐Luk, Bonnie, Choi, Jong Seob, Swaby, Carter, Luk, Matthew, Feller, Laine, Ghosh, Baishakhi, Niederkofler, Austin, Tieng, Ethan, Sherman, Ethan, Chen, Daniel, Upadya, Nisha, Zhang, Rachel, Kim, Deok‐Ho, and Sidhaye, Venkataramana
- Subjects
MICROPHYSIOLOGICAL systems ,LUNG diseases ,CIGARETTE smoke ,SMOKING ,CHRONIC obstructive pulmonary disease ,LUNGS - Abstract
The interactions between immune cells and epithelial cells influence the progression of many respiratory diseases, such as chronic obstructive pulmonary disease (COPD). In vitro models allow for the examination of cells in controlled environments. However, these models lack the complex 3D architecture and vast multicellular interactions between the lung resident cells and infiltrating immune cells that can mediate cellular response to insults. In this study, three complementary microphysiological systems are presented to delineate the effects of cigarette smoke and respiratory disease on the lung epithelium. First, the Transwell system allows the co‐culture of pulmonary immune and epithelial cells to evaluate cellular and monolayer phenotypic changes in response to cigarette smoke exposure. Next, the human and mouse precision‐cut lung slices system provides a physiologically relevant model to study the effects of chronic insults like cigarette smoke with the dissection of specific interaction of immune cell subtypes within the structurally complex tissue environment. Finally, the lung‐on‐a‐chip model provides an adaptable system for live imaging of polarized epithelial tissues that mimic the in vivo environment of the airways. Using a combination of these models, a complementary approach is provided to better address the intricate mechanisms of lung disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Influence of intersignaling crosstalk on the intracellular localization of YAP/TAZ in lung cells
- Author
-
Govorova, I. A., Nikitochkina, S. Y., and Vorotelyak, E. A.
- Published
- 2024
- Full Text
- View/download PDF
4. CALU‐3 lung cells three‐dimensionally assembled onto CellFate® matrix present angiotensin‐converting enzyme‐2 activity.
- Author
-
dos Santos, Jeniffer Farias, dos Reis, Emily Marques, Berti, Fernanda Vieira, Colla, Guilherme, Koepp, Janice, and Nunes, Viviane Abreu
- Abstract
Currently, there is a great need for the development of three‐dimensional (3D) in vitro lung models. Particularly, the production of a suitable 3D model of pulmonary epithelium for understanding the pathophysiology of diseases such as the COVID‐19 must consider the tissue architecture and presence, for example, of the angiotensin‐converting enzyme‐2 (ACE‐2) in the cells. Different polymeric membranes are being used to support cell culturing, especially of lung cells, however, there is still no information about the culture of these cells onto bacterial nanocellulose (BNC) matrices. We have used the BNC matrix CellFate® as a support for the assembly of a 3D in vitro model of lung epithelium, composed of human lung fibroblasts (HLF) and lung adenocarcinoma cells (CALU‐3). CellFate® matrices were made from bacterial fermentation resulting in a natural and biocompatible biopolymer. Cells were cultured onto CellFate® and maintained in a 5% CO2 humidified atmosphere at 37°C. Cell viability was assessed by the resazurin method The samples were, then, exposed to the air–liquid interface (ALI), and histologically analyzed. ACE‐2 activity was verified on the hydrolyze of the fluorogenic substrate Mca‐APK(Dnp)‐OH, and its presence was evaluated by flow cytometry. The expression of the anionic transporter SLCO3A1 was evaluated by qPCR. Cell viability analysis indicates that CellFate® was not toxic to these cells. By flow cytometry, the presence of the ACE‐2 was identified in the CALU‐3 cells surface corroborating the results obtained from enzymatic activity analysis. The SLCO3A1 transporter expression was identified in cells cultured onto CellFate®, but not in cells cultured onto the transwell (control). CALU‐3 cells cultivated onto CellFate® resulted in a pseudostratified organization, a typical morphology of the human respiratory tract epithelium. The current model opens perspectives for studies involving physiological characterization, improving its relevance for the understanding of the pathophysiology of diseases as well as the response to drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Human Lung Organoids—A Novel Experimental and Precision Medicine Approach.
- Author
-
Kühl, Laura, Graichen, Pauline, von Daacke, Nele, Mende, Anne, Wygrecka, Malgorzata, Potaczek, Daniel P., Miethe, Sarah, and Garn, Holger
- Subjects
- *
LUNGS , *INDUCED pluripotent stem cells , *INDIVIDUALIZED medicine , *EXPERIMENTAL medicine , *ORGANOIDS , *CELL culture , *GLOBAL burden of disease - Abstract
The global burden of respiratory diseases is very high and still on the rise, prompting the need for accurate models for basic and translational research. Several model systems are currently available ranging from simple airway cell cultures to complex tissue-engineered lungs. In recent years, human lung organoids have been established as highly transferrable three-dimensional in vitro model systems for lung research. For acute infectious and chronic inflammatory diseases as well as lung cancer, human lung organoids have opened possibilities for precise in vitro research and a deeper understanding of mechanisms underlying lung injury and regeneration. Human lung organoids from induced pluripotent stem cells or from adult stem cells of patients' samples introduce tools for understanding developmental processes and personalized medicine approaches. When further state-of-the-art technologies and protocols come into use, the full potential of human lung organoids can be harnessed. High-throughput assays in drug development, gene therapy, and organoid transplantation are current applications of organoids in translational research. In this review, we emphasize novel approaches in translational and personalized medicine in lung research focusing on the use of human lung organoids. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Redox-Dependent Activation of Lung Epithelial STAT3 Is Required for Inducible Protection against Bacterial Pneumonia.
- Author
-
Kulkarni, Vikram V., Yongxing Wang, Garcia, Jezreel Pantaleon, and Evans, Scott E.
- Abstract
The lung epithelium is dynamic, capable of considerable structural and functional plasticity in response to pathogen challenges. Our laboratory has demonstrated that an inhaled combination of a Toll-like receptor (TLR) 2/6 agonist and a TLR9 agonist (Pam2ODN) results in robust protection against otherwise lethal pneumonias. We have previously shown that intact epithelial TLR signaling and generation of multisource epithelial reactive oxygen species (ROS) are required for inducible protection. Further investigating the mechanisms underlying this phenomenon of inducible resistance, reverse-phase protein array analysis demonstrated robust STAT3 (signal transducer and activator of transcription 3) phosphorylation following treatment of lung epithelial cells. We show here that Pam2ODN-induced STAT3 phosphorylation is IL-6–independent. We further found that therapeutic epithelial STAT3 activation is required for inducible protection against Pseudomonas aeruginosa pneumonia. Additional studies showed that inhibiting epithelial dual oxidases or scavenging ROS significantly reduced the Pam2ODN induction of STAT3 phosphorylation, suggesting a proximal role for ROS in inducible STAT3 activation. Dissecting these mechanisms, we analyzed the contributions of redox-sensitive kinases and found that Pam2ODN activated epithelial growth factor receptor in an ROS-dependent manner that is required for therapeutically inducible STAT3 activation. Taken together, we demonstrate that epithelial STAT3 is imperative for Pam2ODN’s function and describe a novel redox-based mechanism for its activation. These key mechanistic insights may facilitate strategies to leverage inducible epithelial resistance to protect susceptible patients during periods of peak vulnerability. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Benzo[a]pyrene induces NLRP1 expression and promotes prolonged inflammasome signaling.
- Author
-
Risa Kohno, Yuka Nagata, Tomohiro Ishihara, Chisato Amma, Yayoi Inomata, Takafumi Seto, and Ryo Suzuki
- Subjects
INFLAMMASOMES ,ARYL hydrocarbon receptors ,PYRENE ,POLYCYCLIC aromatic hydrocarbons ,REACTIVE oxygen species - Abstract
Benzo[a]pyrene (BaP), a polycyclic aromatic hydrocarbon in the air, triggers pulmonary inflammation. This study focused on BaP-induced inflammation in the alveolar epithelium. A549 cells were stimulated with BaP for four days. BaP treatment markedly increased NLRP1 expression but slightly decreased NLRP3. Furthermore, aryl hydrocarbon receptor (AhR) knockdown displayed no increase in BaP-induced NLRP1 expression. Similar results were also observed by blocking reactive oxygen species (ROS), which is mediated through AhR, suggesting that the AhR-ROS axis operates in BaP-induced NLRP1 expression. p53 involvement in ROS-mediated NLRP1 induction has also been implied. When we confirmed inflammasome activation in cells treated with BaP for four days, while BaP transiently activated NLRP3, it predominantly activated the NLRP1 inflammasome. These findings have led to the conclusion that BaP could be a potential ligand for the NLRP1 inflammasome persistently observed in the lung epithelium. Our study may provide additional evidence for the sustained pulmonary inflammation caused by environmental air pollution. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
8. Targeting the lung epithelium after intravenous delivery by directed evolution of underexplored sites on the AAV capsid
- Author
-
David Goertsen, Nick Goeden, Nicholas C. Flytzanis, and Viviana Gradinaru
- Subjects
AAV ,vector engineering ,directed evolution ,gene delivery ,lung ,lung epithelium ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Advances in adeno-associated virus (AAV) engineering have provided exciting new tools for research and potential solutions for gene therapy. However, the lung has not received the same tailored engineering as other major targets of debilitating genetic disorders. To address this, here we engineered the surface-exposed residues AA452-458 of AAV9 capsid proteins at the three-fold axis of symmetry and employed a Cre-transgenic-based screening platform to identify AAV capsids targeted to the lung after intravenous delivery in mice. Using a custom image processing pipeline to quantify transgene expression across whole tissue images, we found that one engineered variant, AAV9.452sub.LUNG1, displays dramatically improved transgene expression in lung tissue after systemic delivery in mice. This improved transduction extends to alveolar epithelial type II cells, expanding the toolbox for gene therapy research for diseases specific to the lung.
- Published
- 2022
- Full Text
- View/download PDF
9. Effects of hypoxia on bronchial and alveolar epithelial cells linked to pathogenesis in chronic lung disorders.
- Author
-
Berggren-Nylund, Rebecca, Ryde, Martin, Löfdahl, Anna, Ibáñez-Fonseca, Arturo, Kåredal, Monica, Westergren-Thorsson, Gunilla, Tufvesson, Ellen, and Larsson-Callerfelt, Anna-Karin
- Subjects
VASCULAR endothelial growth factors ,EPITHELIAL cells ,FIBROBLAST growth factors ,TRANSFORMING growth factors ,EPIDERMAL growth factor - Abstract
Introduction: Chronic lung disorders involve pathological alterations in the lung tissue with hypoxia as a consequence. Hypoxia may influence the release of inflammatory mediators and growth factors including vascular endothelial growth factor (VEGF) and prostaglandin (PG)E
2 . The aim of this work was to investigate how hypoxia affects human lung epithelial cells in combination with profibrotic stimuli and its correlation to pathogenesis. Methods: Human bronchial (BEAS-2B) and alveolar (hAELVi) epithelial cells were exposed to either hypoxia (1% O2 ) or normoxia (21% O2 ) during 24 h, with or without transforming growth factor (TGF)-β1. mRNA expression of genes and proteins related to disease pathology were analysed with qPCR, ELISA or immunocytochemistry. Alterations in cell viability and metabolic activity were determined. Results: In BEAS-2B and hAELVi, hypoxia significantly dowregulated genes related to fibrosis, mitochondrial stress, oxidative stress, apoptosis and inflammation whereas VEGF receptor 2 increased. Hypoxia increased the expression of Tenascin-C, whereas both hypoxia and TGF-β1 stimuli increased the release of VEGF, IL-6, IL-8 and MCP-1 in BEAS-2B. In hAELVi, hypoxia reduced the release of fibroblast growth factor, epidermal growth factor, PGE2, IL-6 and IL-8, whereas TGF-β1 stimulus significantly increased the release of PGE2 and IL-6. TGF-β1 stimulated BEAS-2B cells showed a decreased release of VEGF-A and IL-8, while TGF-β1 stimulated hAELVi cells showed a decreased release of PGE2 and IL-8 during hypoxia compared to normoxia. Metabolic activity was significantly increased by hypoxia in both epithelial cell types. Discussion: In conclusion, our data indicate that bronchial and alveolar epithelial cells respond differently to hypoxia and profibrotic stimuli. The bronchial epithelium appears more responsive to changes in oxygen levels and remodelling processes compared to the alveoli, suggesting that hypoxia may be a driver of pathogenesis in chronic lung disorders. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
10. Innate immune responses in pneumonia.
- Author
-
Korkmaz, Filiz T. and Traber, Katrina E.
- Subjects
IMMUNE response ,NATURAL immunity ,PNEUMONIA ,PARTICULATE matter ,STROMAL cells - Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Differential Gene Expression Induced by Different TLR Agonists in A549 Lung Epithelial Cells Is Modulated by CRISPR Activation of TLR10.
- Author
-
Knez, Špela, Narat, Mojca, and Ogorevc, Jernej
- Subjects
- *
EPITHELIAL cells , *GENE expression , *ANTIMICROBIAL peptides , *CRISPRS , *TOLL-like receptors , *LUNGS - Abstract
Toll-like receptor 10 (TLR10) is the only member of the TLR family whose function and ligand have not been clearly described. Literature reports on its function are contradictory and suggest a possible immunomodulatory role that depends on the cell type, the pathogen, and the level of TLR10 expression. To investigate the regulatory role of TLR10 in A549 lung epithelial cells, we overexpressed TLR10 using CRISPRa technology and examined the differential expression of various genes involved in TLR signaling activated by different TLR ligands, namely dsRNA, LPS, and Pam3Cys. The expression of proinflammatory cytokines, such as IL1β, IFNβ, TNFα, IL8, CXCL10, and CCL20, decreased in the challenged cells overexpressing TLR10, whereas the expression of the anti-inflammatory cytokine IL10 and the antimicrobial peptide hβD-2 increased. For several of the regulated inflammatory markers, we were able to show the change in gene expression was translated to the protein level. It appears that TLR10 can function as an anti-inflammatory in A549 cells, depending on its expression level and that the mode of action may be virulence factor-specific. The potential suppression of inflammation by regulating expression of TLR10 in lung epithelial cells may allow the development of new approaches to balance an inflammatory response and prevent extensive tissue damage in respiratory diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. iNOS Deletion in Alveolar Epithelium Cannot Reverse the Elastase-Induced Emphysema in Mice.
- Author
-
Gredic, Marija, Sharma, Vinita, Hadzic, Stefan, Wu, Cheng-Yu, Pak, Oleg, Kojonazarov, Baktybek, Duerr, Julia, Mall, Marcus A., Guenther, Andreas, Schermuly, Ralph T., Grimminger, Friedrich, Seeger, Werner, Kraut, Simone, Sommer, Natascha, and Weissmann, Norbert
- Subjects
- *
LUNGS , *CHRONIC obstructive pulmonary disease , *NITRIC-oxide synthases , *SYSTOLIC blood pressure , *PULMONARY hypertension , *CHRONIC bronchitis - Abstract
Background: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. In addition to chronic bronchitis and emphysema, patients often develop at least mild pulmonary hypertension (PH). We previously demonstrated that inhibition of inducible nitric oxide synthase (iNOS) prevents and reverses emphysema and PH in mice. Interestingly, strong iNOS upregulation was found in alveolar epithelial type II cells (AECII) in emphysematous murine lungs, and peroxynitrite, which can be formed from iNOS-derived NO, was shown to induce AECII apoptosis in vitro. However, the specific cell type(s) that drive(s) iNOS-dependent lung regeneration in emphysema/PH has (have) not been identified yet. Aim: we tested whether iNOS knockout in AECII affects established elastase-induced emphysema in mice. Methods: four weeks after a single intratracheal instillation of porcine pancreatic elastase for the induction of emphysema and PH, we induced iNOS knockout in AECII in mice, and gave an additional twelve weeks for the potential recovery. Results: iNOS knockout in AECII did not reduce elastase-induced functional and structural lung changes such as increased lung compliance, decreased mean linear intercept and increased airspace, decreased right ventricular function, increased right ventricular systolic pressure and increased pulmonary vascular muscularization. In vitro, iNOS inhibition did not reduce apoptosis of AECII following exposure to a noxious stimulus. Conclusion: taken together, our data demonstrate that iNOS deletion in AECII is not sufficient for the regeneration of emphysematous murine lungs, and suggest that iNOS expression in pulmonary vascular or stromal cells might be critically important in this regard. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Advanced epithelial lung and gut barrier models demonstrate passage of microplastic particles
- Author
-
Joanne M. Donkers, Elena M. Höppener, Ilya Grigoriev, Lena Will, Barbro N. Melgert, Bas van der Zaan, Evita van de Steeg, and Ingeborg M. Kooter
- Subjects
Microplastics ,In vitro ,Membrane passage ,Lung epithelium ,Intestinal tissue ,Environmental pollution ,TD172-193.5 ,Polymers and polymer manufacture ,TP1080-1185 - Abstract
Abstract Micro- and nanoplastics (MNP) can be found virtually everywhere around us in the biosphere and food chain, therefore humans are continuously exposed to MNP, mainly via inhalation and ingestion. Here, we have applied physiologically relevant human-based advanced in vitro models representing the lung (MucilAir™) and gut (InTESTine™ and Intestinal Explant Barrier Chip (IEBC)) to study membrane passage of various MNP and their potency to induce cytotoxic effects, barrier disturbances or pro-inflammatory cell activation. Selected MNP of various materials (polystyrene, polyethylene, nylon, car tire, and marine MNP collected from the ocean), shapes (spheres, fragments and fibers), sizes (0.05–100 μm), some of which were fluorescently labelled for tracking, were included. Without affecting cell viability, nylon fibers and (cleaned) HDPE (high density polyethylene) disrupted the MucilAir™ epithelial barrier. Luminal exposure to polystyrene particles (1 and 10 μm) and pristine HDPE fragments significantly decreased human colon tissue functionality. Furthermore, all polystyrene particles (0.05, 1 and 10 μm) affected tissue viability in porcine jejunum, ileum and colon tissue after 5 h exposure, and this was further confirmed in the IEBC after 24 h of exposure to 10 μm polystyrene particles and nylon fibers. Exposure to nylon fibers and its supernatant led to pro-inflammatory cell activation, as shown by increased IL-6 release in MucilAir™ and in human colon tissue after 96 or 24 h, respectively. Regarding transepithelial penetration of the MNP, permeability of 0.05 μm polystyrene spheres in the MucilAir™ lung cell model reached 3.6 ± 1.2% after 24 h. With 3.37 ± 0.46% after 5 h under static conditions and 5.5 ± 1.3% after 24 h under microfluidic conditions MNP permeability across intestinal tissue was highest for the largest (10 μm) polystyrene spheres. Confocal microscopy confirmed the translocation of MNP across the lung and intestinal epithelial barrier. In conclusion, we present a study revealing the passage of MNP over the epithelium of advanced in vitro models for the lung and intestine barrier. Furthermore, pro-inflammatory cell activation and disrupted barrier integrity were observed after exposure to several of the tested MNP. Future research is needed to further identify the effects of shape, size and material on these processes and subsequently the health effects of humans.
- Published
- 2022
- Full Text
- View/download PDF
14. Single-Cell Transcriptomic Profiling of Pluripotent Stem Cell-Derived SCGB3A2+ Airway Epithelium
- Author
-
McCauley, Katherine B, Alysandratos, Konstantinos-Dionysios, Jacob, Anjali, Hawkins, Finn, Caballero, Ignacio S, Vedaie, Marall, Yang, Wenli, Slovik, Katherine J, Morley, Michael, Carraro, Gianni, Kook, Seunghyi, Guttentag, Susan H, Stripp, Barry R, Morrisey, Edward E, and Kotton, Darrell N
- Subjects
Lung ,Stem Cell Research - Induced Pluripotent Stem Cell ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Embryonic - Human ,Respiratory ,Animals ,Cell Differentiation ,Cell Line ,Cell Lineage ,Cell Plasticity ,Epithelium ,Gene Expression Profiling ,Genes ,Reporter ,Humans ,Induced Pluripotent Stem Cells ,Kinetics ,Mice ,Secretoglobins ,Sequence Analysis ,RNA ,Single-Cell Analysis ,Solubility ,Spheroids ,Cellular ,Time Factors ,Transcriptome ,Wnt Signaling Pathway ,airway ,alveoli ,directed differentiation ,lung epithelium ,pluripotent stem cells ,single-cell RNA sequencing ,Biochemistry and Cell Biology ,Clinical Sciences - Abstract
Lung epithelial lineages have been difficult to maintain in pure form in vitro, and lineage-specific reporters have proven invaluable for monitoring their emergence from cultured pluripotent stem cells (PSCs). However, reporter constructs for tracking proximal airway lineages generated from PSCs have not been previously available, limiting the characterization of these cells. Here, we engineer mouse and human PSC lines carrying airway secretory lineage reporters that facilitate the tracking, purification, and profiling of this lung subtype. Through bulk and single-cell-based global transcriptomic profiling, we find PSC-derived airway secretory cells are susceptible to phenotypic plasticity exemplified by the tendency to co-express both a proximal airway secretory program as well as an alveolar type 2 cell program, which can be minimized by inhibiting endogenous Wnt signaling. Our results provide global profiles of engineered lung cell fates, a guide for improving their directed differentiation, and a human model of the developing airway.
- Published
- 2018
15. The pulmonary effects of STAT3 deficiency.
- Author
-
Gilje, Elizabeth A. and Abbott, Jordan K.
- Published
- 2023
- Full Text
- View/download PDF
16. Tight Junctions, the Epithelial Barrier, and Toll-like Receptor-4 During Lung Injury.
- Author
-
Godbole, Nachiket M., Chowdhury, Asif Alam, Chataut, Neha, and Awasthi, Shanjana
- Subjects
- *
TIGHT junctions , *LUNG injuries , *WATER-electrolyte balance (Physiology) , *EPITHELIAL cells , *PNEUMONIA - Abstract
Lung epithelium is constantly exposed to the environment and is critically important for the orchestration of initial responses to infectious organisms, toxins, and allergic stimuli, and maintenance of normal gaseous exchange and pulmonary function. The integrity of lung epithelium, fluid balance, and transport of molecules is dictated by the tight junctions (TJs). The TJs are formed between adjacent cells. We have focused on the topic of the TJ structure and function in lung epithelial cells. This review includes a summary of the last twenty years of literature reports published on the disrupted TJs and epithelial barrier in various lung conditions and expression and regulation of specific TJ proteins against pathogenic stimuli. We discuss the molecular signaling and crosstalk among signaling pathways that control the TJ structure and function. The Toll-like receptor-4 (TLR4) recognizes the pathogen- and damage-associated molecular patterns released during lung injury and inflammation and coordinates cellular responses. The molecular aspects of TLR4 signaling in the context of TJs or the epithelial barrier are not fully known. We describe the current knowledge and possible networking of the TLR4-signaling with cellular and molecular mechanisms of TJs, lung epithelial barrier function, and resistance to treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Development of an In Vitro Model of SARS-CoV-Induced Acute Lung Injury for Studying New Therapeutic Approaches.
- Author
-
Shevtsova, Yulia A., Goryunov, Kirill V., Babenko, Valentina A., Pevzner, Irina B., Vtorushina, Valentina V., Inviyaeva, Evgeniya V., Krechetova, Lyubov V., Zorova, Ljubava D., Plotnikov, Egor Y., Zorov, Dmitry B., Sukhikh, Gennady T., and Silachev, Denis N.
- Subjects
LUNGS ,THERAPEUTICS ,ETIOLOGY of diseases ,POLYMYXIN B ,MONONUCLEAR leukocytes ,LIPOPOLYSACCHARIDES ,MULTIPOTENT stem cells - Abstract
One of the causes of death of patients infected by SARS-CoV-2 is the induced respiratory failure caused by excessive activation of the immune system, the so-called "cytokine storm", leading to damage to lung tissue. In vitro models reproducing various stages of the disease can be used to explore the pathogenetic mechanisms and therapeutic approaches to treating the consequences of a cytokine storm. We have developed an in vitro test system for simulating damage to the pulmonary epithelium as a result of the development of a hyperinflammatory reaction based on the co-cultivation of pulmonary epithelial cells (A549 cells) and human peripheral blood mononuclear cells (PBMC) primed with lipopolysaccharide (LPS). In this model, after 24 h of co-cultivation, a sharp decrease in the rate of proliferation of A549 cells associated with the intrinsic development of oxidative stress and, ultimately, with the induction of PANoptotic death were observed. There was a significant increase in the concentration of 40 cytokines/chemokines in a conditioned medium, including TNF-α, IFN-α, IL-6, and IL-1a, which corresponded to the cytokine profile in patients with severe manifestation of COVID-19. In order to verify the model, the analysis of the anti-inflammatory effects of well-known substances (dexamethasone, LPS from Rhodobacter sphaeroides (LPS-RS), polymyxin B), as well as multipotent mesenchymal stem cells (MSC) and MSC-derived extracellular vesicles (EVs) was carried out. Dexamethasone and polymyxin B restored the proliferative activity of A549 cells and reduced the concentration of proinflammatory cytokines. MSC demonstrated an ambivalent effect through stimulated production of both pro-inflammatory cytokines and growth factors that regenerate lung tissue. LPS-RS and EVs showed no significant effect. The developed test system can be used to study molecular and cellular pathological processes and to evaluate the effectiveness of various therapeutic approaches for the correction of hyperinflammatory response in COVID-19 patients. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
18. Long-term alterations in lung epithelial cells after EL-RSV infection exacerbate allergic responses through IL-1β-induced pathways.
- Author
-
Morris SB, Ocadiz-Ruiz R, Asai N, Malinczak CA, Rasky AJ, Lombardo GK, Velarde EM, Ptaschinski C, Zemans RL, Lukacs NW, and Fonseca W
- Subjects
- Animals, Mice, Signal Transduction, Disease Models, Animal, Humans, Alveolar Epithelial Cells metabolism, Alveolar Epithelial Cells immunology, Female, Epigenesis, Genetic, Hypersensitivity immunology, Hypersensitivity etiology, Allergens immunology, Epithelial Cells immunology, Epithelial Cells metabolism, Mice, Inbred BALB C, Respiratory Syncytial Virus Infections immunology, Interleukin-1beta metabolism, Respiratory Syncytial Viruses, Asthma immunology, Asthma etiology, Interleukin-33 metabolism, Interleukin-33 genetics, Lung immunology
- Abstract
Early-life (EL) respiratory infections increase pulmonary disease risk, especially EL-Respiratory Syncytial Virus (EL-RSV) infections linked to asthma. Mechanisms underlying asthma predisposition remain unknown. In this study, we examined the long-term effects on the lung after four weeks post EL-RSV infection. We identified alterations in the lung epithelial cell, with a rise in the percentage of alveolar type 2 epithelial cells (AT2) and a decreased percentage of cells in the AT1 and AT2-AT1 subclusters, as well as upregulation of Bmp2 and Krt8 genes that are associated with AT2-AT1 trans-differentiation, suggesting potential defects in lung repair processes. We identified persistent upregulation of asthma-associated genes, including Il33. EL-RSV-infected mice allergen-challenged exhibited exacerbated allergic response, with significant upregulation of Il33 in the lung and AT2 cells. Similar long-term effects were observed in mice exposed to EL-IL-1β. Notably, treatment with IL-1ra during acute EL-RSV infection mitigated the long-term alveolar alterations and the allergen-exacerbated response. Finally, epigenetic modifications in the promoter of the Il33 gene were detected in AT2 cells harvested from EL-RSV and EL-IL1β groups, suggesting that long-term alteration in the epithelium after RSV infection is dependent on the IL-1β pathway. This study provides insight into the molecular mechanisms of asthma predisposition after RSV infection., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. Human Lung Organoids—A Novel Experimental and Precision Medicine Approach
- Author
-
Laura Kühl, Pauline Graichen, Nele von Daacke, Anne Mende, Malgorzata Wygrecka, Daniel P. Potaczek, Sarah Miethe, and Holger Garn
- Subjects
disease models ,lung epithelium ,airways ,organoids ,personalized medicine ,translational research ,Cytology ,QH573-671 - Abstract
The global burden of respiratory diseases is very high and still on the rise, prompting the need for accurate models for basic and translational research. Several model systems are currently available ranging from simple airway cell cultures to complex tissue-engineered lungs. In recent years, human lung organoids have been established as highly transferrable three-dimensional in vitro model systems for lung research. For acute infectious and chronic inflammatory diseases as well as lung cancer, human lung organoids have opened possibilities for precise in vitro research and a deeper understanding of mechanisms underlying lung injury and regeneration. Human lung organoids from induced pluripotent stem cells or from adult stem cells of patients’ samples introduce tools for understanding developmental processes and personalized medicine approaches. When further state-of-the-art technologies and protocols come into use, the full potential of human lung organoids can be harnessed. High-throughput assays in drug development, gene therapy, and organoid transplantation are current applications of organoids in translational research. In this review, we emphasize novel approaches in translational and personalized medicine in lung research focusing on the use of human lung organoids.
- Published
- 2023
- Full Text
- View/download PDF
20. Type IV collagen drives alveolar epithelial–endothelial association and the morphogenetic movements of septation
- Author
-
Loscertales, Maria, Nicolaou, Fotini, Jeanne, Marion, Longoni, Mauro, Gould, Douglas B, Sun, Yunwei, Maalouf, Faouzi I, Nagy, Nandor, and Donahoe, Patricia K
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Nonembryonic - Non-Human ,Stem Cell Research ,Genetics ,Underpinning research ,2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,A549 Cells ,Animals ,Basement Membrane ,Cell Differentiation ,Cell Movement ,Cell Proliferation ,Cells ,Cultured ,Chick Embryo ,Coculture Techniques ,Collagen Type IV ,Endothelial Cells ,Epithelial Cells ,Fibroblasts ,Humans ,Lung ,Mice ,Mice ,Knockout ,Microarray Analysis ,Morphogenesis ,Mutation ,Myofibroblasts ,Peptide Fragments ,Up-Regulation ,Type IV collagen ,Basement membrane ,Blood-gas barrier ,Alveolar development ,Lung epithelium ,Lung vasculature ,Alveolar myofibroblast migration and differentiation ,Blood–gas barrier ,Developmental Biology ,Biological sciences - Abstract
BackgroundType IV collagen is the main component of the basement membrane that gives strength to the blood-gas barrier (BGB). In mammals, the formation of a mature BGB occurs primarily after birth during alveologenesis and requires the formation of septa from the walls of the saccule. In contrast, in avians, the formation of the BGB occurs rapidly and prior to hatching. Mutation in basement membrane components results in an abnormal alveolar phenotype; however, the specific role of type IV collagen in regulating alveologenesis remains unknown.ResultsWe have performed a microarray expression analysis in late chick lung development and found that COL4A1 and COL4A2 were among the most significantly upregulated genes during the formation of the avian BGB. Using mouse models, we discovered that mutations in murine Col4a1 and Col4a2 genes affected the balance between lung epithelial progenitors and differentiated cells. Mutations in Col4a1 derived from the vascular component were sufficient to cause defects in vascular development and the BGB. We also show that Col4a1 and Col4a2 mutants displayed disrupted myofibroblast proliferation, differentiation and migration. Lastly, we revealed that addition of type IV collagen protein induced myofibroblast proliferation and migration in monolayer culture and increased the formation of mesenchymal-epithelial septal-like structures in co-culture.ConclusionsOur study showed that type IV collagen and, therefore the basement membrane, play fundamental roles in coordinating alveolar morphogenesis. In addition to its role in the formation of epithelium and vasculature, type IV collagen appears to be key for alveolar myofibroblast development by inducing their proliferation, differentiation and migration throughout the developing septum.
- Published
- 2016
21. Two-hybrid screening of FAM13A protein partners in lung epithelial cells
- Author
-
Manon Ruffin, Kristin E. Thompson, Harriet Corvol, and Loic Guillot
- Subjects
FAM13A ,Chronic lung diseases ,Lung epithelium ,Two-hybrid screening ,Medicine ,Biology (General) ,QH301-705.5 ,Science (General) ,Q1-390 - Abstract
Abstract Objectives Family with sequence similarity 13 member A (FAM13A) genetic variants have been associated with several chronic respiratory diseases including chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF) and lung cancer. The FAM13A protein includes a RhoGTPase activating protein (RhoGAP) domain known to participate in various cellular mechanisms including cell proliferation. While intensive genomic studies have been performed to reveal its involvement in lung diseases, the biological role of FAM13A protein is still not completely elucidated. Results We therefore performed a two-hybrid screening to identify protein partners of FAM13A using a human lung cancer cDNA library. We identified several protein partners with a high confidence score. Researchers in the field of chronic lung diseases may benefit from this two-hybrid screening data which may reveal new research pathways to decipher.
- Published
- 2020
- Full Text
- View/download PDF
22. Differential Gene Expression Induced by Different TLR Agonists in A549 Lung Epithelial Cells Is Modulated by CRISPR Activation of TLR10
- Author
-
Špela Knez, Mojca Narat, and Jernej Ogorevc
- Subjects
CRISPR/dCas9 ,cytokines ,inflammation ,TLR10 ,lung epithelium ,innate immunity ,Microbiology ,QR1-502 - Abstract
Toll-like receptor 10 (TLR10) is the only member of the TLR family whose function and ligand have not been clearly described. Literature reports on its function are contradictory and suggest a possible immunomodulatory role that depends on the cell type, the pathogen, and the level of TLR10 expression. To investigate the regulatory role of TLR10 in A549 lung epithelial cells, we overexpressed TLR10 using CRISPRa technology and examined the differential expression of various genes involved in TLR signaling activated by different TLR ligands, namely dsRNA, LPS, and Pam3Cys. The expression of proinflammatory cytokines, such as IL1β, IFNβ, TNFα, IL8, CXCL10, and CCL20, decreased in the challenged cells overexpressing TLR10, whereas the expression of the anti-inflammatory cytokine IL10 and the antimicrobial peptide hβD-2 increased. For several of the regulated inflammatory markers, we were able to show the change in gene expression was translated to the protein level. It appears that TLR10 can function as an anti-inflammatory in A549 cells, depending on its expression level and that the mode of action may be virulence factor-specific. The potential suppression of inflammation by regulating expression of TLR10 in lung epithelial cells may allow the development of new approaches to balance an inflammatory response and prevent extensive tissue damage in respiratory diseases.
- Published
- 2022
- Full Text
- View/download PDF
23. iNOS Deletion in Alveolar Epithelium Cannot Reverse the Elastase-Induced Emphysema in Mice
- Author
-
Marija Gredic, Vinita Sharma, Stefan Hadzic, Cheng-Yu Wu, Oleg Pak, Baktybek Kojonazarov, Julia Duerr, Marcus A. Mall, Andreas Guenther, Ralph T. Schermuly, Friedrich Grimminger, Werner Seeger, Simone Kraut, Natascha Sommer, and Norbert Weissmann
- Subjects
COPD ,emphysema ,iNOS ,lung epithelium ,AECII ,Cytology ,QH573-671 - Abstract
Background: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. In addition to chronic bronchitis and emphysema, patients often develop at least mild pulmonary hypertension (PH). We previously demonstrated that inhibition of inducible nitric oxide synthase (iNOS) prevents and reverses emphysema and PH in mice. Interestingly, strong iNOS upregulation was found in alveolar epithelial type II cells (AECII) in emphysematous murine lungs, and peroxynitrite, which can be formed from iNOS-derived NO, was shown to induce AECII apoptosis in vitro. However, the specific cell type(s) that drive(s) iNOS-dependent lung regeneration in emphysema/PH has (have) not been identified yet. Aim: we tested whether iNOS knockout in AECII affects established elastase-induced emphysema in mice. Methods: four weeks after a single intratracheal instillation of porcine pancreatic elastase for the induction of emphysema and PH, we induced iNOS knockout in AECII in mice, and gave an additional twelve weeks for the potential recovery. Results: iNOS knockout in AECII did not reduce elastase-induced functional and structural lung changes such as increased lung compliance, decreased mean linear intercept and increased airspace, decreased right ventricular function, increased right ventricular systolic pressure and increased pulmonary vascular muscularization. In vitro, iNOS inhibition did not reduce apoptosis of AECII following exposure to a noxious stimulus. Conclusion: taken together, our data demonstrate that iNOS deletion in AECII is not sufficient for the regeneration of emphysematous murine lungs, and suggest that iNOS expression in pulmonary vascular or stromal cells might be critically important in this regard.
- Published
- 2022
- Full Text
- View/download PDF
24. SARS-CoV-2 activates lung epithelial cell proinflammatory signaling and leads to immune dysregulation in COVID-19 patients
- Author
-
Huarong Chen, Weixin Liu, Yifei Wang, Dabin Liu, Liuyang Zhao, and Jun Yu
- Subjects
COVID-19 ,SARS-CoV-2 ,scRNA-seq ,lung epithelium ,Dysregulated immune response ,Medicine ,Medicine (General) ,R5-920 - Abstract
Background: The outbreak of Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection has become a global health emergency. We aim to decipher SARS-CoV-2 infected cell types, the consequent host immune response and their interplay in lung of COVID-19 patients. Methods: We analyzed single-cell RNA sequencing (scRNA-seq) data of bronchoalveolar lavage fluid (BALF) samples from 10 healthy donors, 6 severe COVID-19 patients and 3 mild recovered patients. The expressions of SARS-CoV-2 receptors (ACE2 and TMPRSS2) were examined among different cell types. The immune cells infiltration patterns, their expression profiles, and interplays between immune cells and SARS-CoV-2 target cells were further investigated. Findings: Compared to healthy controls, ACE2 and TMPRSS2 expressions were significantly higher in lung epithelial cells of COVID-19 patients, in particular club and ciliated cells. SARS-CoV-2 activated pro-inflammatory genes and interferon/cytokine signaling in these cells. In severe COVID-19 patients, significantly higher neutrophil, but lower macrophage in lung was observed along with markedly increased cytokines expression compared with healthy controls and mild patients. By contrast, neutrophil and macrophage returned to normal level whilst more T and NK cells accumulation were observed in mild patients. Moreover, SARS-CoV-2 infection altered the community interplays of lung epithelial and immune cells: interactions between the club and immune cells were higher in COVID-19 patients compared to healthy donors; on the other hand, immune-immune cells interactions appeared the strongest in mild patients. Interpretation: SARS-CoV-2 could infect lung epithelium, alter communication patterns between lung epithelial cells and immune system, and drive dysregulated host immune response in COVID-19 patients.
- Published
- 2021
- Full Text
- View/download PDF
25. The development of an in vitro 3D model of goblet cell hyperplasia using MUC5AC expression and repeated whole aerosol exposures.
- Author
-
Haswell, Linsey E., Smart, David, Jaunky, Tomasz, Baxter, Andrew, Santopietro, Simone, Meredith, Stuart, Camacho, Oscar M., Breheny, Damien, Thorne, David, and Gaca, Marianna D.
- Subjects
- *
AEROSOLS , *CIGARETTE smoke , *SMOKING , *TOBACCO products , *HYPERPLASIA - Abstract
• Highlights importance of repeat exposure assays for modelling disease pathogenesis. • Details the development and challenges of complex repeat exposure studies. • First publication of repeat whole smoke exposure with increase in MUC5AC expression. • MUC5AC increase is a clinically relevant endpoint for smoking related disease. • Comparison of repeat tobacco heated product exposures with air and cigarette smoke. Goblet cell hyperplasia and overproduction of airway mucin are characteristic features of the lung epithelium of smokers and COPD patients. Tobacco heating products (THPs) are a potentially less risky alternative to combustible cigarettes, and through continued use solus THPs may reduce smoking-related disease risk. Using the MucilAir™ in vitro lung model, a 6-week feasibility study was conducted investigating the effect of repeated cigarette smoke (1R6F), THP aerosol and air exposure. Tissues were exposed to nicotine-matched whole aerosol doses 3 times/week. Endpoints assessed were dosimetry, tight-junction integrity, cilia beat frequency (CBF) and active area (AA), cytokine secretion and airway mucin MUC5AC expression. Comparison of incubator and air exposed controls indicated exposures did not have a significant effect on the transepithelial electrical resistance (TEER), CBF and AA of the tissues. Cytokine secretion indicated clear differences in secretion patterns in response to 1R6F and THP exposure. 1R6F exposure resulted in a significant decrease in the TEER and AA (p=0.000 and p=0.000, respectively), and an increase in MUC5AC positive cells (p=0.002). Repeated THP exposure did not result in a significant change in MUC5AC positive cells. This study demonstrates repeated cigarette smoke whole aerosol exposure can induce these morphological changes in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. Role of bone marrow-derived mesenchymal stem cells in alleviating pulmonary epithelium damage and extracellular matrix remodeling in a rat model of lung fibrosis induced by amiodarone.
- Author
-
Abdel Halim, Alyaa S., Ahmed, Hanaa H., Aglan, Hadeer A., Abdel Hamid, Fatma F., and Mohamed, Mohamed R.
- Subjects
- *
MESENCHYMAL stem cells , *CONNECTIVE tissue growth factor , *EXTRACELLULAR matrix , *ANIMAL disease models , *PULMONARY fibrosis , *LUNGS - Abstract
The therapeutic role of mesenchymal stem cells (MSCs) in cases of amiodarone (AD) induced pulmonary fibrosis (PF) has not been well studied. Also, the period required by MSCs to attain full therapeutic effectiveness has not yet been assessed. We investigated the potential curative effect of bone marrow-derived MSCs (BM-MSCs) and conditioned media (CM) from BM-MSCs on AD induced PF by focusing on pulmonary epithelium injury and repair, and extracellular matrix (ECM) remodeling. We used 64 Wistar rats divided into eight groups: negative control group; PF group; three PF groups treated with BM-MSCs for 1, 2 or 4 months; and three PF groups treated with CM for 1, 2 and 4 months. Serum levels of Clara cell secretory protein (CC16) and keratinocyte growth factor (KGF) were measured. Gene expression of type I collagen (COL1A1) and connective tissue growth factor (CTGF) was evaluated in pulmonary tissue. Treatment of PF challenged rats with BM-MSCs or CM caused reduced CC16 levels, increased KGF levels, reduced expression of COL1A1 and CTGF, histological improvement following lung injury, and decreased collagen accumulation. Treatment with BM-MSCs exhibited greater amelioration of PF than CM. BM-MSCs or CM treatment for 2 and 4 months exhibited better resolution of fibrosis than treatment for 1 month. BM-MSCs are promising for treating PF due to their attenuation of ECM deposition in addition to alleviating pulmonary epithelium damage and initiating its repair. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
27. The common ABCA3E292V variant disrupts AT2 cell quality control and increases susceptibility to lung injury and aberrant remodeling.
- Author
-
Tomer, Yaniv, Wambach, Jennifer, Knudsen, Lars, Ming Zhao, Rodriguez, Luis R., Murthy, Aditi, White, Frances V., Venosa, Alessandro, Katzen, Jeremy, Ochs, Matthias, Hamvas, Aaron, Beers, Michael F., and Mulugeta, Surafel
- Subjects
- *
LUNG injuries , *RESPIRATORY distress syndrome , *GREEN fluorescent protein , *LUNGS , *QUALITY control , *PULMONARY fibrosis - Abstract
ATP-binding cassette class A3 (ABCA3) is a lipid transporter that plays a critical role in pulmonary surfactant function. The substitution of valine for glutamic acid at codon 292 (E292V) produces a hypomorphic variant that accounts for a significant portion of ABCA3 mutations associated with lung disorders spanning from neonatal respiratory distress syndrome and childhood interstitial lung disease to diffuse parenchymal lung disease (DPLD) in adults including pulmonary fibrosis. The mechanisms by which this and similar ABCA3 mutations disrupt alveolar type 2 (AT2) cell homeostasis and cause DPLD are largely unclear. The present study, informed by a patient homozygous for the E292V variant, used an in vitro and a preclinical murine model to evaluate the mechanisms by which E292V expression promotes aberrant lung injury and parenchymal remodeling. Cell lines stably expressing enhanced green fluorescent protein (EGFP)-tagged ABCA3 isoforms show a functional deficiency of the ABCA3E292V variant as a lipid transporter. AT2 cells isolated from mice constitutively homozygous for ABCA3E292V demonstrate the presence of small electron-dense lamellar bodies, time-dependent alterations in macroautophagy, and induction of apoptosis. These changes in AT2 cell homeostasis are accompanied by a spontaneous lung phenotype consisting of both age-dependent inflammation and fibrillary collagen deposition in alveolar septa. Older ABCA3E292V mice exhibit increased vulnerability to exogenous lung injury by bleomycin. Collectively, these findings support the hypothesis that the ABCA3E292V variant is a susceptibility factor for lung injury through effects on surfactant deficiency and impaired AT2 cell autophagy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
28. Development of an In Vitro Model of SARS-CoV-Induced Acute Lung Injury for Studying New Therapeutic Approaches
- Author
-
Yulia A. Shevtsova, Kirill V. Goryunov, Valentina A. Babenko, Irina B. Pevzner, Valentina V. Vtorushina, Evgeniya V. Inviyaeva, Lyubov V. Krechetova, Ljubava D. Zorova, Egor Y. Plotnikov, Dmitry B. Zorov, Gennady T. Sukhikh, and Denis N. Silachev
- Subjects
COVID-19 ,cytokine storm ,model ,lung epithelium ,oxidative stress ,Therapeutics. Pharmacology ,RM1-950 - Abstract
One of the causes of death of patients infected by SARS-CoV-2 is the induced respiratory failure caused by excessive activation of the immune system, the so-called “cytokine storm”, leading to damage to lung tissue. In vitro models reproducing various stages of the disease can be used to explore the pathogenetic mechanisms and therapeutic approaches to treating the consequences of a cytokine storm. We have developed an in vitro test system for simulating damage to the pulmonary epithelium as a result of the development of a hyperinflammatory reaction based on the co-cultivation of pulmonary epithelial cells (A549 cells) and human peripheral blood mononuclear cells (PBMC) primed with lipopolysaccharide (LPS). In this model, after 24 h of co-cultivation, a sharp decrease in the rate of proliferation of A549 cells associated with the intrinsic development of oxidative stress and, ultimately, with the induction of PANoptotic death were observed. There was a significant increase in the concentration of 40 cytokines/chemokines in a conditioned medium, including TNF-α, IFN-α, IL-6, and IL-1a, which corresponded to the cytokine profile in patients with severe manifestation of COVID-19. In order to verify the model, the analysis of the anti-inflammatory effects of well-known substances (dexamethasone, LPS from Rhodobacter sphaeroides (LPS-RS), polymyxin B), as well as multipotent mesenchymal stem cells (MSC) and MSC-derived extracellular vesicles (EVs) was carried out. Dexamethasone and polymyxin B restored the proliferative activity of A549 cells and reduced the concentration of proinflammatory cytokines. MSC demonstrated an ambivalent effect through stimulated production of both pro-inflammatory cytokines and growth factors that regenerate lung tissue. LPS-RS and EVs showed no significant effect. The developed test system can be used to study molecular and cellular pathological processes and to evaluate the effectiveness of various therapeutic approaches for the correction of hyperinflammatory response in COVID-19 patients.
- Published
- 2022
- Full Text
- View/download PDF
29. Induction of alveolar and bronchiolar phenotypes in human lung organoids
- Author
-
Laurence Hoareau, Agnete S. T. Engelsen, Marianne Aanerud, Maria Paula Ramnefjell, Pirjo‐Riitta Salminen, Fabian Gärtner, Thomas Halvorsen, Helge Ræder, and Mariann H. L. Bentsen
- Subjects
3D model ,lung epithelium ,organoids ,Physiology ,QP1-981 - Abstract
Abstract Patient‐derived organoids have revolutionized biomedical research and therapies by "transferring the patient into the Petri dish". In vitro access to human lung organoids representing distal lung tissue, i.e. alveolar organoids, would facilitate research pertaining to a wide range of medical conditions and might open for a future approach to individualized treatment.We propose a protocol to derive a single human lung biopsy towards both alveolar and bronchiolar organoids. By modulating Wnt pathway, we obtained a differential gene expression of the main markers for both subtypes, such as a higher expression of surfactant protein C in alveolar organoids or a higher expression of mucine 5AC in bronchiolar organoids. Although the specific cell enrichment was not complete, the differentiation was observed as early as passage 1 based on morphology, and confirmed by QPCR and histology at passage 2. These results are consistent with a functional specification of lung epithelium towards both alveoli‐ and bronchi‐enriched organoids from first passages
- Published
- 2021
- Full Text
- View/download PDF
30. The critical role of collagen VI in lung development and chronic lung disease
- Author
-
Jared A. Mereness and Thomas J. Mariani
- Subjects
Collagen VI ,Lung development ,Chronic lung disease ,Extracellular matrix ,Basement membrane ,Lung epithelium ,Biology (General) ,QH301-705.5 - Abstract
Type VI collagen (collagen VI) is an obligate extracellular matrix component found mainly in the basement membrane region of many mammalian tissues and organs, including skeletal muscle and throughout the respiratory system. Collagen VI is probably most recognized in medicine as the genetic cause of a spectrum of muscular dystrophies, including Ullrich Congenital Myopathy and Bethlem Myopathy. Collagen VI is thought to contribute to myopathy, at least in part, by mediating muscle fiber integrity by anchoring myoblasts to the muscle basement membrane. Interestingly, collagen VI myopathies present with restrictive respiratory insufficiency, thought to be due primarily to thoracic muscular weakening. Although it was recently recognized as one of the (if not the) most abundant collagens in the mammalian lung, there is a substantive knowledge gap concerning its role in respiratory system development and function. A few studies have suggested that collagen VI insufficiency is associated with airway epithelial cell survival and altered lung function. Our recent work suggested collagen VI may be a genomic risk factor for chronic lung disease in premature infants. Using this as motivation, we thoroughly assessed the role of collagen VI in lung development and in lung epithelial cell biology. Here, we describe the state-of-the-art for collagen VI cell and developmental biology within the respiratory system, and reveal its essential roles in normal developmental processes and airway epithelial cell phenotype and intracellular signaling.
- Published
- 2021
- Full Text
- View/download PDF
31. Induction of alveolar and bronchiolar phenotypes in human lung organoids.
- Author
-
Hoareau, Laurence, Engelsen, Agnete S. T., Aanerud, Marianne, Ramnefjell, Maria Paula, Salminen, Pirjo‐Riitta, Gärtner, Fabian, Halvorsen, Thomas, Ræder, Helge, and Bentsen, Mariann H. L.
- Subjects
- *
HUMAN phenotype , *ORGANOIDS , *PROTEIN C , *LUNGS , *PROTEIN expression - Abstract
Patient‐derived organoids have revolutionized biomedical research and therapies by "transferring the patient into the Petri dish". In vitro access to human lung organoids representing distal lung tissue, i.e. alveolar organoids, would facilitate research pertaining to a wide range of medical conditions and might open for a future approach to individualized treatment.We propose a protocol to derive a single human lung biopsy towards both alveolar and bronchiolar organoids. By modulating Wnt pathway, we obtained a differential gene expression of the main markers for both subtypes, such as a higher expression of surfactant protein C in alveolar organoids or a higher expression of mucine 5AC in bronchiolar organoids. Although the specific cell enrichment was not complete, the differentiation was observed as early as passage 1 based on morphology, and confirmed by QPCR and histology at passage 2. These results are consistent with a functional specification of lung epithelium towards both alveoli‐ and bronchi‐enriched organoids from first passages [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. AP-3-dependent targeting of flippase ATP8A1 to lamellar bodies suppresses activation of YAP in alveolar epithelial type 2 cells.
- Author
-
Kook, Seunghyi, Ping Wang, Shufang Meng, Jetter, Christopher S., Sucre, Jennifer M. S., Benjamin, John T., Gokey, Jason J., Hanby, Hayley A., Jaume, Alexa, Goetzl, Laura, Marks, Michael S., and Guttentag, Susan H.
- Subjects
- *
PULMONARY fibrosis , *CELL migration , *HOMEOSTASIS , *ENDOSOMES , *ADENOSINE triphosphatase , *COMMERCIAL products - Abstract
Lamellar bodies (LBs) are lysosome-related organelles (LROs) of surfactant-producing alveolar type 2 (AT2) cells of the distal lung epithelium. Trafficking pathways to LBs have been understudied but are likely critical to AT2 cell homeostasis given associations between genetic defects of endosome to LRO trafficking and pulmonary fibrosis in Hermansky Pudlak syndrome (HPS). Our prior studies uncovered a role for AP-3, defective in HPS type 2, in trafficking Peroxiredoxin-6 to LBs. We now show that the P4-type ATPase ATP8A1 is sorted by AP-3 from early endosomes to LBs through recognition of a C-terminal dileucine-based signal. Disruption of the AP-3/ATP8A1 interaction causes ATP8A1 accumulation in early sorting and/or recycling endosomes, enhancing phosphatidylserine exposure on the cytosolic leaflet. This in turn promotes activation of Yes-activating protein, a transcriptional coactivator, augmenting cell migration and AT2 cell numbers. Together, these studies illuminate a mechanism whereby loss of AP-3-mediated trafficking contributes to a toxic gain-of-function that results in enhanced and sustained activation of a repair pathway associated with pulmonary fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. Alterations in oxygen metabolism are associated to lung toxicity triggered by silver nanoparticles exposure.
- Author
-
Garcés, Mariana, Magnani, Natalia D, Pecorelli, Alessandra, Calabró, Valeria, Marchini, Timoteo, Cáceres, Lourdes, Pambianchi, Erika, Galdoporpora, Juan, Vico, Tamara, Salgueiro, Jimena, Zubillaga, Marcela, Moretton, Marcela A, Desimone, Martin F, Alvarez, Silvia, Valacchi, Giuseppe, and Evelson, Pablo
- Subjects
- *
SILVER nanoparticles , *LUNGS , *RESPIRATORY organs , *REACTIVE oxygen species , *NADPH oxidase , *NANOPARTICLE toxicity , *PULMONARY toxicology , *OXYGEN metabolism - Abstract
Along with the AgNP applications development, the concern about their possible toxicity has increasingly gained attention. As the respiratory system is one of the main exposure routes, the aim of this study was to evaluate the harmful effects developed in the lung after an acute AgNP exposure. In vivo studies using Balb/c mice intranasally instilled with 0.1 mg AgNP/kg b.w, were performed. 99mTc-AgNP showed the lung as the main organ of deposition, where, in turn, AgNP may exert barrier injury observed by increased protein content and total cell count in BAL samples. In vivo acute exposure showed altered lung tissue O 2 consumption due to increased mitochondrial active respiration and NOX activity. Both O 2 consumption processes release ROS triggering the antioxidant system as observed by the increased SOD, catalase and GPx activities and a decreased GSH/GSSG ratio. In addition, increased protein oxidation was observed after AgNP exposure. In A549 cells, exposure to 2.5 μg/mL AgNP during 1 h resulted in augment NOX activity, decreased mitochondrial ATP associated respiration and higher H 2 O 2 production rate. Lung 3D tissue model showed AgNP-initiated barrier alterations as TEER values decreased and morphological alterations. Taken together, these results show that AgNP exposure alters O 2 metabolism leading to alterations in oxygen metabolism lung toxicity. AgNP-triggered oxidative damage may be responsible for the impaired lung function observed due to alveolar epithelial injury. [Display omitted] • Exposure to AgNP altered lung tissue O 2 consumption due to increased mitochondrial active respiration and NOX activity. • Increased NOX activity and mitochondrial H 2 O 2 production rate led to oxidative damage. • Lung 3D tissue model showed AgNP-initiated barrier alterations as TEER values decreased. • Oxidative damage may be responsible for the impaired lung function observed due to alveolar epithelial injury. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
34. In vitro Alternatives to Acute Inhalation Toxicity Studies in Animal Models—A Perspective
- Author
-
Dania Movia, Solene Bruni-Favier, and Adriele Prina-Mello
- Subjects
toxicity testing alternatives ,inhalation studies ,In vitro alternatives ,air-liquid interface (ALI) culture ,lung epithelium ,Biotechnology ,TP248.13-248.65 - Abstract
When assessing the risk and hazard of a non-pharmaceutical compound, the first step is determining acute toxicity, including toxicity following inhalation. Inhalation is a major exposure route for humans, and the respiratory epithelium is the first tissue that inhaled substances directly interact with. Acute inhalation toxicity testing for regulatory purposes is currently performed only in rats and/or mice according to OECD TG403, TG436, and TG433 test guidelines. Such tests are biased by the differences in the respiratory tract architecture and function across species, making it difficult to draw conclusions on the potential hazard of inhaled compounds in humans. Research efforts have been therefore focused on developing alternative, human-relevant models, with emphasis on the creation of advanced In vitro models. To date, there is no In vitro model that has been accepted by regulatory agencies as a stand-alone replacement for inhalation toxicity testing in animals. Here, we provide a brief introduction to current OECD test guidelines for acute inhalation toxicity, the interspecies differences affecting the predictive value of such tests, and the current regulatory efforts to advance alternative approaches to animal-based inhalation toxicity studies. We then list the steps that should allow overcoming the current challenges in validating In vitro alternatives for the successful replacement of animal-based inhalation toxicity studies. These steps are inclusive and descriptive, and should be detailed when adopting in house-produced 3D cell models for inhalation tests. Hence, we provide a checklist of key parameters that should be reported in any future scientific publications for reproducibility and transparency.
- Published
- 2020
- Full Text
- View/download PDF
35. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche
- Author
-
N. Valerio Dorrello and Gordana Vunjak-Novakovic
- Subjects
lung ,bioengineering ,regeneration ,lung vasculature ,lung epithelium ,lung support ex vivo ,Biotechnology ,TP248.13-248.65 - Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
- Published
- 2020
- Full Text
- View/download PDF
36. Mechanisms of Epithelial Immunity Evasion by Respiratory Bacterial Pathogens
- Author
-
Lokesh Sharma, Jingjing Feng, Clemente J. Britto, and Charles S. Dela Cruz
- Subjects
bacterial dissemination ,host pathogen interaction ,epithelial immunity ,pathogen evolution ,lung epithelium ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Bacterial lung infections are major healthcare challenges killing millions of people worldwide and resulting in a huge economic burden. Both basic and clinical research have elucidated host mechanisms that contribute to the bacterial clearance where an indispensable role of immune cells has been established. However, the role of respiratory epithelial cells in bacterial clearance has garnered limited attention due to their weak inflammatory or phagocytic ability compared to immune cells such as macrophages and neutrophils. These studies often underappreciate the fact that epithelial cells are the most abundant cells in the lung, not only serving as building blocks but also providing immune protection throughout the lung. Epithelial cells function either independently to eradicate the pathogen or communicate with immune cells to orchestrate pathogen clearance. The epithelial cells have multiple mechanisms that include mucus production, antimicrobial peptide production, muco-ciliary clearance, and phagocytosis, all of which contribute to their direct antibacterial function. Secretion of cytokines to recruit immune cells and potentiate their antimicrobial activities is a pathway by which the epithelium contributes to bacterial clearance. Successful pathogens outsmart epithelial resistance and find a way to replicate in sufficient numbers to establish infections in the airway or lung epithelial surfaces. In this mini-review, we discuss evidences that establish important roles for epithelial host defense against invading respiratory bacterial pathogens and demonstrate how pathogens outsmart these epithelial immune mechanisms to successfully establish infection. Finally, we discuss briefly how to boost epithelial immunity to improve outcomes in bacterial lung infections.
- Published
- 2020
- Full Text
- View/download PDF
37. Human Induced Pluripotent Stem Cell-Derived Lung Epithelial System for SARS-CoV-2 Infection Modeling and Its Potential in Drug Repurposing.
- Author
-
Surendran, Harshini, Nandakumar, Swapna, and Pal, Rajarshi
- Subjects
- *
PLURIPOTENT stem cells , *SARS-CoV-2 , *EPITHELIAL cells , *ADULT respiratory distress syndrome , *COVID-19 , *PHARMACOLOGY , *LUNGS - Abstract
The lung is the most vulnerable target for the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, and respiratory failure causing acute respiratory distress syndrome is its foremost outcome. However, the current primary in vitro models in use for SARS-CoV-2 display apparent limitations for modeling such complex human respiratory disease. Although patient cells can directly model the effects of a drug, their availability and capacity for expansion are limited compared with transformed/immortalized cells or tumor-derived cell lines. An additional caveat is that the latter may harbor genetic and metabolic abnormalities making them unsuitable for drug screening. Therefore, it is important to create physiologically relevant human-cell models that can replicate the pathophysiology of SARS-CoV-2, thus facilitating drug testing. In this study, we show preliminary data on how human induced pluripotent stem cells-derived lung epithelial cell system could emerge as a relevant and sensitive platform for modeling SARS-CoV-2 infection and drug screening. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
38. Contribution of mast cells in irritant-induced airway epithelial barrier impairment in vitro.
- Author
-
Van Den Broucke, Sofie, Vanoirbeek, Jeroen, Alfaro-Moreno, Ernesto, and Hoet, Peter
- Subjects
- *
MAST cells , *THYMIC stromal lymphopoietin , *EPITHELIAL cells , *INTERLEUKIN-6 , *GRAPHENE oxide - Abstract
The airway epithelium is continuously exposed to environmental irritants, which can cause adverse effects such as irritant-induced asthma (IIA). Mast cells are located near airway epithelia and are able to respond to a variety of stimuli. We aimed to investigate whether mast cells influence the response of the epithelium upon irritant exposure. Two cell lines and three different seeding conditions, that is, bronchial epithelial cells (16HBE) only, 16HBE with mast cells (HMC-1's) basolaterally, and 16HBE with HMC-1's apically, were established. Upon exposure to the environmental irritants, graphene (G), graphene oxide (GO), diesel exhaust particles (DEPs) or hypochlorite (ClO−), transepithelial electrical resistance (TEER) and paracellular flux of fluorescent-labeled dextrans were determined, along with the release of mediators. Identical experiments were conducted with the Ca2+ ionophore ionomycin. Exposure to G and GO induced a significant and permanent decrease of approximately 70% in TEER after 3 h of exposure, whereas DEP and ClO− exposure resulted in a transient decrease of approximately 20% in TEER. This response pattern was similar in all the different seeding conditions. After 24 h of exposure, fluorescein isothiocyanate–dextran transport was 10-fold greater for G and 5-fold greater for GO in each of the tested seeding conditions, while DEP and ClO− induced no change compared to the control. Upon exposure to the irritants, 16HBE did not release thymic stromal lymphopoietin, interleukin 33 (IL-33), or IL-1α, and HMC-1 cells did not release histamine, IL-6, or IL-8. Epithelial barrier integrity upon treatment with ionomycin was not affected by the presence of HMC-1 cells. A limited amount of IL-6 and IL-8 was released by ionomycin-exposed HMC-1 cells. To conclude, we found that the studied environmental irritants do not directly or indirectly activate HMC-1 cells. These mast cells did not influence the epithelial barrier function upon environmental exposure, and thus currently do not provide additional information for the underlying mechanism of IIA. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
39. First contact: the role of respiratory cilia in host-pathogen interactions in the airways.
- Author
-
Li Eon Kuek and Lee, Robert J.
- Subjects
- *
MUCOCILIARY system , *CILIA & ciliary motion , *RESPIRATORY infections , *CHRONICALLY ill , *EPITHELIAL cells , *ESCALATORS - Abstract
Respiratory cilia are the driving force of the mucociliary escalator, working in conjunction with secreted airway mucus to clear inhaled debris and pathogens from the conducting airways. Respiratory cilia are also one of the first contact points between host and inhaled pathogens. Impaired ciliary function is a common pathological feature in patients with chronic airway diseases, increasing susceptibility to respiratory infections. Common respiratory pathogens, including viruses, bacteria, and fungi, have been shown to target cilia and/or ciliated airway epithelial cells, resulting in a disruption of mucociliary clearance that may facilitate host infection. Despite being an integral component of airway innate immunity, the role of respiratory cilia and their clinical significance during airway infections are still poorly understood. This review examines the expression, structure, and function of respiratory cilia during pathogenic infection of the airways. This review also discusses specific known points of interaction of bacteria, fungi, and viruses with respiratory cilia function. The emerging biological functions of motile cilia relating to intracellular signaling and their potential immunoregulatory roles during infection will also be discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
40. Evaluation and live monitoring of pH-responsive HSA-ZnO nanoparticles using a lung-on-a-chip model.
- Author
-
Meghani, Nileshkumar, Kim, Kyung Hwan, Kim, Soo Hwan, Lee, Sang Ho, and Choi, Kyung Hyun
- Abstract
One of the key problems that have hindered the development and approval of anticancer nanoparticle drug delivery systems is the limited predictability of 2D cell culture and animal models. Here, we describe a biomimetic alveolus-epithelium-on-a-chip (AEOC) model with in-built sensors for monitoring and evaluating pH-responsive zinc oxide quantum dots (QDs)-loaded human serum albumin nanoparticles. This AEOC model closely represents the cancerous alveolus epithelium, which comprises lung cancer cells, as well as stromal cells, such as fibroblasts along with extracellular matrix (ECM) in the form of collagen. ZnO QDs were encapsulated in the HSA nanoparticles with a diameter of 60 nm. The physicochemical properties, quantum dots release, in vitro cytotoxicity, and cellular uptake of HSA-ZnO were evaluated. HSA-ZnO showed higher ZnO loading and encapsulation efficacy. TEER and pH sensors were used to monitor the cells over three days, and real-time data with and without nanoparticle treatment were obtained. Cell viability after treatment with 10 and 50 µg/mL of HSA-ZnO nanoparticles and confocal imaging data confirmed the significant internalization of the nanoparticles under co-culture cellular conditions in the AEOC model. Our designed organ-on-a-chip model has potentially expanded the capabilities of cell culture in biomimetic conditions, and therefore, can provide a low-cost alternative to expensive and tedious animal models for the evaluation of nanomedicines. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
41. Two-hybrid screening of FAM13A protein partners in lung epithelial cells.
- Author
-
Ruffin, Manon, Thompson, Kristin E., Corvol, Harriet, and Guillot, Loic
- Subjects
- *
OBSTRUCTIVE lung diseases , *EPITHELIAL cells , *IDIOPATHIC pulmonary fibrosis , *ANTISENSE DNA , *LUNGS , *LUNG diseases , *CYSTIC fibrosis - Abstract
Objectives: Family with sequence similarity 13 member A (FAM13A) genetic variants have been associated with several chronic respiratory diseases including chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF) and lung cancer. The FAM13A protein includes a RhoGTPase activating protein (RhoGAP) domain known to participate in various cellular mechanisms including cell proliferation. While intensive genomic studies have been performed to reveal its involvement in lung diseases, the biological role of FAM13A protein is still not completely elucidated. Results: We therefore performed a two-hybrid screening to identify protein partners of FAM13A using a human lung cancer cDNA library. We identified several protein partners with a high confidence score. Researchers in the field of chronic lung diseases may benefit from this two-hybrid screening data which may reveal new research pathways to decipher. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
42. Role of the co-transcriptional regulators Yap/Taz in the normal and fibrotic lung epithelia
- Author
-
Alsafadi, Hani N. and Alsafadi, Hani N.
- Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal disease that exhibits patterns of usual interstitial pneumonia with honeycombing. IPF is characterized by damaged distal lung epithelium with excessive tissue scarring and extracellular matrix remodeling. The etiology of IPF is unknown and current therapies cannot end or reverse disease progression. Aberrant reactivation of developmental pathways is evident in IPF. Among these developmental actors are the co-transcriptional regulators Yap and Taz (YT). YT modulate processes such as proliferation, differentiation, and organ size and are regulated by the Hippo pathway. YT do not have a DNA binding domain but act through interaction with other transcription factors (TFs). YT play a role in fibrotic fibroblasts, but their role is not yet known in the fibrotic lung epithelium. The aim of this thesis project is to develop the tools needed to explore the role of Hippo-YT in fibrotic lung epithelium and to identify the TFs that YT interact with to exert their various functions.We first developed a method to simultaneously isolate proximal and distal lung progenitor cells from an individual mouse with the aid of a 3D printed surgical guide and found that the precision of dissecting the lung lobes affects the purity of the isolated distal progenitors and how they behave in organoid assays. We further found the Hippo pathway to be dysregulated in the fibrotic lung epithelium which led to increases in nuclear YT as well as known downstream targets. Interestingly, we found epithelial YT signaling to be actively involved in extracellular matrix remodeling in the fibrotic lung epithelium through modulation of lysyl oxidase expression, a collagen crosslinking enzyme. Targeting YT in vivo using an FDA approved drug ameliorated the fibrotic phenotype, indicating that YT targeting may be an option to treat fibrosis. We further used cleavage under target and release using nuclease (CUT&RUN) to identify the exact motif sequences on the geno
- Published
- 2023
43. Novel Role for CFTR in Fluid Absorption from the Distal Airspaces of the Lung
- Author
-
Fang, X, Fukuda, N, Barbry, P, Sartori, C, Verkman, AS, and Matthay, MA
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Rare Diseases ,Lung ,Cystic Fibrosis ,Respiratory ,Absorption ,Animals ,Bronchodilator Agents ,Chloride Channels ,Cystic Fibrosis Transmembrane Conductance Regulator ,Humans ,In Vitro Techniques ,Isotonic Solutions ,Mice ,Mice ,Inbred C3H ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Pulmonary Edema ,Sodium Chloride ,pulmonary edema ,cystic fibrosis ,lung epithelium ,cAMP ,lung fluid balance ,Physiology ,Biochemistry and cell biology ,Zoology ,Medical physiology - Abstract
The active absorption of fluid from the airspaces of the lung is important for the resolution of clinical pulmonary edema. Although ENaC channels provide a major route for Na(+) absorption, the route of Cl(-) transport has been unclear. We applied a series of complementary approaches to define the role of Cl(-) transport in fluid clearance in the distal airspaces of the intact mouse lung, using wild-type and cystic fibrosis Delta F508 mice. Initial studies in wild-type mice showed marked inhibition of fluid clearance by Cl(-) channel inhibitors and Cl(-) ion substitution, providing evidence for a transcellular route for Cl(-) transport. In response to cAMP stimulation by isoproterenol, clearance was inhibited by the CFTR inhibitor glibenclamide in both wild-type mice and the normal human lung. Although isoproterenol markedly increased fluid absorption in wild-type mice, there was no effect in Delta F508 mice. Radioisotopic clearance studies done at 23 degrees C (to block active fluid absorption) showed approximately 20% clearance of (22)Na in 30 min both without and with isoproterenol. However, the clearance of (36)Cl was increased by 47% by isoproterenol in wild-type mice but was not changed in Delta F508 mice, providing independent evidence for involvement of CFTR in cAMP-stimulated Cl(-) transport. Further, CFTR played a major role in fluid clearance in a mouse model of acute volume-overload pulmonary edema. After infusion of saline (40% body weight), the lung wet-to-dry weight ratio increased by 28% in wild-type versus 64% in Delta F508 mice. These results provide direct evidence for a functionally important role for CFTR in the distal airspaces of the lung.
- Published
- 2002
44. Advanced epithelial lung and gut barrier models demonstrate passage of microplastic particles
- Author
-
Donkers, Joanne M., Höppener, Elena M., Grigoriev, Ilya, Will, Lena, Melgert, Barbro N., van der Zaan, Bas, van de Steeg, Evita, and Kooter, Ingeborg M.
- Published
- 2022
- Full Text
- View/download PDF
45. Biomechanical Response of Lung Epithelial Cells to Iron Oxide and Titanium Dioxide Nanoparticles
- Author
-
Vinícius Rosa Oliveira, Juan José Uriarte, Bryan Falcones, Ignasi Jorba, Walter Araujo Zin, Ramon Farré, Daniel Navajas, and Isaac Almendros
- Subjects
air pollution ,lung epithelium ,cell biomechanics ,nanoparticles ,actomyosin fibers ,Physiology ,QP1-981 - Abstract
Increasing evidence shows that lungs can be damaged by inhalation of nanoparticles (NPs) at environmental and occupational settings. Recent findings have associated the exposure to iron oxide (Fe2O3) and titanium dioxide (TiO2) – NPs widely used in biomedical and clinical research – with pulmonary oxidative stress and inflammation. Although changes on cellular mechanics could contribute to pulmonary inflammation, there is no information regarding the effects of Fe2O3 and TiO2 on alveolar epithelial cell biomechanics. The aim was to investigate the NPs-induced biomechanical effects in terms of cell stiffness and traction forces exerted by human alveolar epithelial cells. Cell Young’s modulus (E) measured by atomic force microscopy in alveolar epithelial cells significantly decreased after exposure to Fe2O3 and TiO2 (∼28 and ∼25%, respectively) compared to control conditions. Moreover, both NPs induced a similar reduction in the traction forces exerted by the alveolar epithelial cells in comparison to the control conditions. Accordingly, immunofluorescence images revealed a reduction of actomyosin stress fibers in response to the exposure to NPs. However, no inflammatory response was detected. In conclusion, an acute exposure of epithelial pulmonary cells to Fe2O3 and TiO2 NPs, which was mild since it was non-cytotoxic and did not induce inflammation, modified cell biomechanical properties which could be translated into damage of the epithelial barrier integrity, suggesting that mild environmental inhalation of Fe2O3 and TiO2 NPs could not be innocuous.
- Published
- 2019
- Full Text
- View/download PDF
46. Palmitic Acid--Rich High-Fat Diet Exacerbates Experimental Pulmonary Fibrosis by Modulating Endoplasmic Reticulum Stress.
- Author
-
Chu, Sarah G., Villalba, Julian A., Liang, Xiaoliang, Xiong, Kevin, Tsoyi, Konstantin, Ith, Bonna, Ayaub, Ehab A., Tatituri, Raju V., Byers, Derek E., Fong-Fu Hsu, El-Chemaly, Souheil, Kim, Edy Y., Yuanyuan Shi, and Rosas, Ivan O.
- Subjects
IDIOPATHIC pulmonary fibrosis ,PALMITIC acid ,LUNG injuries ,BLEOMYCIN ,ANTINEOPLASTIC antibiotics - Abstract
The impact of lipotoxicity on the development of lung fibrosis is unclear. Saturated fatty acids, such as palmitic acid (PA), activate endoplasmic reticulum (ER) stress, a cellular stress response associated with the development of idiopathic pulmonary fibrosis (IPF).We tested the hypothesis that PA increases susceptibility to lung epithelial cell death and experimental fibrosis by modulating ER stress. Total liquid chromatography and mass spectrometry were used to measure fatty acid content in IPF lungs. Wild-type mice were fed a high-fat diet (HFD) rich in PA or a standard diet and subjected to bleomycin-induced lung injury. Lung fibrosis was determined by hydroxyproline content. Mouse lung epithelial cells were treated with PA. ER stress and cell death were assessed by Western blotting, TUNEL staining, and cell viability assays. IPF lungs had a higher level of PA compared with controls. Bleomycin-exposed mice fed an HFD had significantly increased pulmonary fibrosis associated with increased cell death and ER stress compared with those fed a standard diet. PA increased apoptosis and activation of the unfolded protein response in lung epithelial cells. This was attenuated by genetic deletion and chemical inhibition of CD36, a fatty acid transporter. In conclusion, consumption of an HFD rich in saturated fat increases susceptibility to lung fibrosis and ER stress, and PA mediates lung epithelial cell death and ER stress via CD36. These findings demonstrate that lipotoxicity may have a significant impact on the development of lung injury and fibrosis by enhancing pro-death ER stress pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
47. Coordinate regulation of ELF5 and EHF at the chr11p13 CF modifier region.
- Author
-
Swahn, Hannah, Sabith Ebron, Jey, Lamar, Kay‐Marie, Yin, Shiyi, Kerschner, Jenny L., NandyMazumdar, Monali, Coppola, Candice, Mendenhall, Eric M., Leir, Shih‐Hsing, and Harris, Ann
- Subjects
TRANSCRIPTION factors ,LUNG diseases ,CYSTIC fibrosis ,EPITHELIAL cells ,EXOCRINE glands ,CANCER cells ,CHROMATIN - Abstract
E74‐like factor 5 (ELF5) and ETS‐homologous factor (EHF) are epithelial selective ETS family transcription factors (TFs) encoded by genes at chr11p13, a region associated with cystic fibrosis (CF) lung disease severity. EHF controls many key processes in lung epithelial function so its regulatory mechanisms are important. Using CRISPR/Cas9 technology, we removed three key cis‐regulatory elements (CREs) from the chr11p13 region and also activated multiple open chromatin sites with CRISPRa in airway epithelial cells. Deletion of the CREs caused subtle changes in chromatin architecture and site‐specific increases in EHF and ELF5. CRISPRa had most effect on ELF5 transcription. ELF5 levels are low in airway cells but higher in LNCaP (prostate) and T47D (breast) cancer cells. ATAC‐seq in these lines revealed novel peaks of open chromatin at the 5' end of chr11p13 associated with an expressed ELF5 gene. Furthermore, 4C‐seq assays identified direct interactions between the active ELF5 promoter and sites within the EHF locus, suggesting coordinate regulation between these TFs. ChIP‐seq for ELF5 in T47D cells revealed ELF5 occupancy within EHF introns 1 and 6, and siRNA‐mediated depletion of ELF5 enhanced EHF expression. These results define a new role for ELF5 in lung epithelial biology. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
48. Biomechanical Response of Lung Epithelial Cells to Iron Oxide and Titanium Dioxide Nanoparticles.
- Author
-
Oliveira, Vinícius Rosa, Uriarte, Juan José, Falcones, Bryan, Jorba, Ignasi, Zin, Walter Araujo, Farré, Ramon, Navajas, Daniel, and Almendros, Isaac
- Subjects
TITANIUM dioxide nanoparticles ,EPITHELIAL cells ,FERRIC oxide ,TITANIUM oxides ,ATOMIC force microscopy - Abstract
Increasing evidence shows that lungs can be damaged by inhalation of nanoparticles (NPs) at environmental and occupational settings. Recent findings have associated the exposure to iron oxide (Fe
2 O3 ) and titanium dioxide (TiO2 ) – NPs widely used in biomedical and clinical research – with pulmonary oxidative stress and inflammation. Although changes on cellular mechanics could contribute to pulmonary inflammation, there is no information regarding the effects of Fe2 O3 and TiO2 on alveolar epithelial cell biomechanics. The aim was to investigate the NPs-induced biomechanical effects in terms of cell stiffness and traction forces exerted by human alveolar epithelial cells. Cell Young's modulus (E) measured by atomic force microscopy in alveolar epithelial cells significantly decreased after exposure to Fe2 O3 and TiO2 (∼28 and ∼25%, respectively) compared to control conditions. Moreover, both NPs induced a similar reduction in the traction forces exerted by the alveolar epithelial cells in comparison to the control conditions. Accordingly, immunofluorescence images revealed a reduction of actomyosin stress fibers in response to the exposure to NPs. However, no inflammatory response was detected. In conclusion, an acute exposure of epithelial pulmonary cells to Fe2 O3 and TiO2 NPs, which was mild since it was non-cytotoxic and did not induce inflammation, modified cell biomechanical properties which could be translated into damage of the epithelial barrier integrity, suggesting that mild environmental inhalation of Fe2 O3 and TiO2 NPs could not be innocuous. [ABSTRACT FROM AUTHOR]- Published
- 2019
- Full Text
- View/download PDF
49. Mechanistic insight into heterogeneity of trans-plasma membrane electron transport in cancer cell types.
- Author
-
Sherman, Harry G., Jovanovic, Carolyn, Abuawad, Alaa, Kim, Dong-Hyun, Collins, Hilary, Dixon, James E., Cavanagh, Robert, Markus, Robert, Stolnik, Snow, and Rawson, Frankie J.
- Subjects
- *
BIOLOGICAL transport , *ELECTRON transport , *CANCER cells , *CHARGE exchange , *VITAMIN C , *OXIDATIVE phosphorylation - Abstract
Trans-plasma membrane electron transfer (tMPET) is a process by which reducing equivalents, either electrons or reductants like ascorbic acid, are exported to the extracellular environment by the cell. TPMET is involved in a number of physiological process and has been hypothesised to play a role in the redox regulation of cancer metabolism. Here, we use a new electrochemical assay to elucidate the 'preference' of cancer cells for different trans tPMET systems. This aids in proving a biochemical framework for the understanding of tPMET role, and for the development of novel tPMET-targeting therapeutics. We have delineated the mechanism of tPMET in 3 lung cancer cell models to show that the external electron transfer is orchestrated by ascorbate mediated shuttling via tPMET. In addition, the cells employ a different, non-shuttling-based mechanism based on direct electron transfer via Dcytb. Results from our investigations indicate that tPMETs are used differently, depending on the cell type. The data generated indicates that tPMETs may play a fundamental role in facilitation of energy reprogramming in malignant cells, whereby tPMETs are utilised to supply the necessary energy requirement when mitochondrial stress occurs. Our findings instruct a deeper understanding of tPMET systems, and show how different cancer cells may preferentially use distinguishable tPMET systems for cellular electron transfer processes. • Lung cancer cell models are shown to use two transplasma membrane electron transport systems (tPMETs) but in different ratios • One tPMET system is an ascorbate mediated system and the second occurs via a membrane bound DcytB • The cells were shown to upregulate external electron transfer in response to mitochondrial depolarisation • This suggest trans-plasma membrane electron transport systems are used to reprogram cellular energy requirements. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
50. Activation of the mTORC1/PGC-1 axis promotes mitochondrial biogenesis and induces cellular senescence in the lung epithelium.
- Author
-
Summer, Ross, Shaghaghi, Hoora, Schriner, DeLeila, Roque, Willy, Sales, Dominic, Cuevas-Mora, Karina, Desai, Vilas, Bhushan, Alok, Ramirez, Maria I., and Romero, Freddy
- Subjects
- *
IDIOPATHIC pulmonary fibrosis , *OBSTRUCTIVE lung diseases , *CANCER treatment , *EPITHELIAL cells , *MITOCHONDRIA formation , *LUNGS - Abstract
Cellular senescence is a biological process by which cells lose their capacity to proliferate yet remain metabolically active. Although originally considered a protective mechanism to limit the formation of cancer, it is now appreciated that cellular senescence also contributes to the development of disease, including common respiratory ailments such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. While many factors have been linked to the development of cellular senescence, mitochondrial dysfunction has emerged as an important causative factor. In this study, we uncovered that the mitochondrial biogenesis pathway driven by the mammalian target of rapamycin/peroxisome proliferator-activated receptor-γ complex 1α/β (mTOR/PGC-1α/β) axis is markedly upregulated in senescent lung epithelial cells. Using two different models, we show that activation of this pathway is associated with other features characteristic of enhanced mitochondrial biogenesis, including elevated number of mitochondrion per cell, increased oxidative phosphorylation, and augmented mitochondrial reactive oxygen species (ROS) production. Furthermore, we found that pharmacological inhibition of the mTORC1 complex with rapamycin not only restored mitochondrial homeostasis but also reduced cellular senescence to bleomycin in lung epithelial cells. Likewise, mitochondrial-specific antioxidant therapy also effectively inhibited mTORC1 activation in these cells while concomitantly reducing mitochondrial biogenesis and cellular senescence. In summary, this study provides a mechanistic link between mitochondrial biogenesis and cellular senescence in lung epithelium and suggests that strategies aimed at blocking the mTORC1/PGC- 1α/β axis or reducing ROS-induced molecular damage could be effective in the treatment of senescence-associated lung diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.