1
|
Sharma M, Huber E, Arnesdotter E, Behrsing HP, Bettmann A, Brandwein D, Constant S, Date R, Deshpande A, Fabian E, Gupta A, Gutierrez R, Gutleb AC, Hargrove MM, Hollings M, Hutter V, Jarabek AM, Kaluzhny Y, Landsiedel R, Milchak L, Moyer RA, Murray JR, Page K, Patel M, Pearson SN, Petersen EJ, Reinke E, Roldan N, Roper C, Scaglione JB, Settivari RS, Stucki AO, Verstraelen S, Wallace JL, McCullough S, Clippinger AJ. Minimum information for reporting on the TEER (trans-epithelial/endothelial electrical resistance) assay (MIRTA). Arch Toxicol 2025; 99:57-66. [PMID: 39365315 PMCID: PMC11742365 DOI: 10.1007/s00204-024-03879-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024]
Abstract
Standard information reporting helps to ensure that assay conditions and data are consistently reported and to facilitate inter-laboratory comparisons. Here, we present recommendations on minimum information for reporting on the TEER (trans-epithelial/endothelial electrical resistance) assay (MIRTA). The TEER assay is extensively used to evaluate the health of an epithelial/endothelial cell culture model and as an indicator of the potential toxicity of a test substance. This publication is the result of an international collaboration─called the RespTox (Respiratory Toxicity) Collaborative─through which twelve laboratories shared their protocols for assessing the barrier function of respiratory epithelial cells using the TEER assay following exposure to substances. The protocols from each laboratory were reviewed to identify general steps for performing the TEER assay, interlaboratory differences between steps, the rationale for differences, whether these differences impact results or cross-laboratory comparisons between TEER measurements. While the MIRTA recommendations are focused on respiratory epithelial cell systems, these recommendations can be adapted for other cell systems that form barriers. The use of these recommendations will support data transparency and reproducibility, reduce challenges in data interpretation, enable cross-laboratory comparisons, help assess study quality, and facilitate the incorporation of the TEER assay into national and international testing guidance.
Collapse
Affiliation(s)
- Monita Sharma
- PETA Science Consortium International e.V., 70499, Stuttgart, Germany.
| | - Erin Huber
- Exposure and Protection, RTI International, 3040 East Cornwallis Road, Durham, NC, USA
| | - Emma Arnesdotter
- Environmental Research and Innovation (ERIN) Department, Luxemburg Institute of Science and Technology, 5 Avenue Des Hauts-Fourneaux, 4362, Esch-Sur-Alzette, Grand Duchy of Luxembourg
| | | | - Adam Bettmann
- PETA Science Consortium International e.V., 70499, Stuttgart, Germany
- , 3M Company, St. Paul, MN, 55144, USA
| | | | - Samuel Constant
- Epithelix Sàrl, Chemin Des Aulx 18, 1228, Plan-Les-Ouates, Switzerland
| | - Rahul Date
- Jai Research Foundation, N. H. 48, Near Daman-Ganga Bridge, Valvada, Gujarat, 396105, India
| | - Abhay Deshpande
- Jai Research Foundation, N. H. 48, Near Daman-Ganga Bridge, Valvada, Gujarat, 396105, India
| | - Eric Fabian
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany
| | - Amit Gupta
- Life Science Research, Battelle Memorial Institute, Columbus, OH, 43201, USA
| | - Robert Gutierrez
- Materials Measurement Laboratory, National Institute of Standards and Technology (NIST), Gaithersburg, MD, 20899, USA
| | - Arno C Gutleb
- Environmental Research and Innovation (ERIN) Department, Luxemburg Institute of Science and Technology, 5 Avenue Des Hauts-Fourneaux, 4362, Esch-Sur-Alzette, Grand Duchy of Luxembourg
| | - Marie M Hargrove
- Syngenta Crop Protection, 410 Swing Rd, Greensboro, NC, 27409, USA
| | - Michael Hollings
- Labcorp Early Development Laboratories Ltd., North Yorkshire, HG3 1PY, UK
| | - Victoria Hutter
- ImmuONE Ltd, Sycamore House, 16 Leyden Road, Stevenage, Herts, SG1 2BP, UK
- Centre for Topical Drug Delivery and Toxicology, University of Hertfordshire, College Lane Campus, Hatfield, Herts, AL10 9AB, UK
| | - Annie M Jarabek
- Center for Public Health and Environmental Assessment (CPHEA), Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, Washington, NC, 27711, USA
| | - Yulia Kaluzhny
- MatTek Life Sciences, Ashland, MA, 01721, USA
- InVitroTox Solutions Consulting, Newton, USA
| | - Robert Landsiedel
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany
- Pharmacy, Pharmacology and Toxicology, Free University of Berlin, Berlin, Germany
| | - Lawrence Milchak
- , 3M Company, St. Paul, MN, 55144, USA
- Kimberly-Clark Corporation, Irving, USA
| | - Robert A Moyer
- Life Science Research, Battelle Memorial Institute, Columbus, OH, 43201, USA
| | - Jessica R Murray
- Center for Public Health and Environmental Assessment (CPHEA), Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, Washington, NC, 27711, USA
| | - Kathryn Page
- The Clorox Company, 4900 Johnson Dr, Pleasanton, CA, 94588, USA
| | - Manish Patel
- Jai Research Foundation, N. H. 48, Near Daman-Ganga Bridge, Valvada, Gujarat, 396105, India
| | - Stephanie N Pearson
- Life Science Research, Battelle Memorial Institute, Columbus, OH, 43201, USA
| | - Elijah J Petersen
- Materials Measurement Laboratory, National Institute of Standards and Technology (NIST), Gaithersburg, MD, 20899, USA
| | | | - Nuria Roldan
- PETA Science Consortium International e.V., 70499, Stuttgart, Germany
| | - Clive Roper
- Roper Toxicology Consulting Limited, Edinburgh, EH3 6AD, UK
| | | | | | - Andreas O Stucki
- PETA Science Consortium International e.V., 70499, Stuttgart, Germany
| | - Sandra Verstraelen
- Environmental Intelligence Unit, Flemish Institute for Technological Research (VITO), 2400, Mol, Belgium
| | - Joanne L Wallace
- Charles River Laboratories Edinburgh Ltd, Elphinstone Research Centre, Tranent, East Lothian, EH33 2NE, UK
| | - Shaun McCullough
- Exposure and Protection, RTI International, 3040 East Cornwallis Road, Durham, NC, USA
| | - Amy J Clippinger
- PETA Science Consortium International e.V., 70499, Stuttgart, Germany
| |
Collapse
|
2
|
Schierz AK, Rößler G, Schneider JP, Tschanz SA, Werlein C, Jonigk DD, Schipke J, Mühlfeld C. Distribution and volume of mitochondria in alveolar epithelial type 1 cells in infant and adult human lungs. Histochem Cell Biol 2024; 163:7. [PMID: 39557665 PMCID: PMC11573795 DOI: 10.1007/s00418-024-02332-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/20/2024]
Abstract
Alveolar epithelial type I (AE1) cells with their wide spatial expansion form approximately 95% of the outer surface area of the air-blood barrier inside the lung. Serial block-face scanning electron microscopy (SBF-SEM) investigations led to the hypothesis that AE1 cell mitochondria are preferentially distributed as aggregates in those parts of AE1 cells that are located above connective tissue pillars between capillaries, thus not increasing the thickness of the diffusion distance for oxygen and carbon dioxide. Furthermore, it was hypothesised that postnatal development requires adapting the amount and distribution of mitochondria in AE1 cells. Human lung samples from three infant (26 and 30 days, 6 months) and three adult (20, 39 and 40 years) samples were investigated by light microscopy, transmission electron microscopy and stereology. The volume fraction of mitochondria was similar in infant and adult lungs with a mean value of 6.3%. The ratio between mitochondrial profiles on top of capillaries or above connective tissue pillars was approximately 3:1 in infants and adults. However, regarding the volume of both cytoplasmic compartments, infants showed a higher number of mitochondrial profiles on top of capillaries while adults showed a higher number above connective tissue pillars. Samples of three additional adult lungs were analysed by confocal laser scanning microscopy. Again, mitochondria were not preferentially found as aggregates above connective tissue pillars. In conclusion, AE1 cell mitochondria were not preferentially found as aggregates, showed the same volume density in infants and adults but differed in distribution between the age groups.
Collapse
Affiliation(s)
- Arne K Schierz
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Giacomo Rößler
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jan Philipp Schneider
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Stefan A Tschanz
- University of Bern, Institute of Anatomy, Baltzerstrasse 2, 3008, Bern, Switzerland
| | - Christopher Werlein
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Hannover Medical School, Institute of Pathology, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Danny D Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Institute of Pathology, University Clinics of RWTH University, Aachen, Germany
| | - Julia Schipke
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Christian Mühlfeld
- Hannover Medical School, Institute of Functional and Applied Anatomy, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany.
| |
Collapse
|
3
|
Silva AR, de Souza e Souza KFC, Souza TBD, Younes-Ibrahim M, Burth P, de Castro Faria Neto HC, Gonçalves-de-Albuquerque CF. The Na/K-ATPase role as a signal transducer in lung inflammation. Front Immunol 2024; 14:1287512. [PMID: 38299144 PMCID: PMC10827986 DOI: 10.3389/fimmu.2023.1287512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is marked by damage to the capillary endothelium and alveolar epithelium following edema formation and cell infiltration. Currently, there are no effective treatments for severe ARDS. Pathologies such as sepsis, pneumonia, fat embolism, and severe trauma may cause ARDS with respiratory failure. The primary mechanism of edema clearance is the epithelial cells' Na/K-ATPase (NKA) activity. NKA is an enzyme that maintains the electrochemical gradient and cell homeostasis by transporting Na+ and K+ ions across the cell membrane. Direct injury on alveolar cells or changes in ion transport caused by infections decreases the NKA activity, loosening tight junctions in epithelial cells and causing edema formation. In addition, NKA acts as a receptor triggering signal transduction in response to the binding of cardiac glycosides. The ouabain (a cardiac glycoside) and oleic acid induce lung injury by targeting NKA. Besides enzymatic inhibition, the NKA triggers intracellular signal transduction, fostering proinflammatory cytokines production and contributing to lung injury. Herein, we reviewed and discussed the crucial role of NKA in edema clearance, lung injury, and intracellular signaling pathway activation leading to lung inflammation, thus putting the NKA as a protagonist in lung injury pathology.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Thamires Bandeira De Souza
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Mauricio Younes-Ibrahim
- Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Perniss A, Boonen B, Tonack S, Thiel M, Poharkar K, Alnouri MW, Keshavarz M, Papadakis T, Wiegand S, Pfeil U, Richter K, Althaus M, Oberwinkler J, Schütz B, Boehm U, Offermanns S, Leinders-Zufall T, Zufall F, Kummer W. A succinate/SUCNR1-brush cell defense program in the tracheal epithelium. SCIENCE ADVANCES 2023; 9:eadg8842. [PMID: 37531421 PMCID: PMC10396310 DOI: 10.1126/sciadv.adg8842] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Host-derived succinate accumulates in the airways during bacterial infection. Here, we show that luminal succinate activates murine tracheal brush (tuft) cells through a signaling cascade involving the succinate receptor 1 (SUCNR1), phospholipase Cβ2, and the cation channel transient receptor potential channel subfamily M member 5 (TRPM5). Stimulated brush cells then trigger a long-range Ca2+ wave spreading radially over the tracheal epithelium through a sequential signaling process. First, brush cells release acetylcholine, which excites nearby cells via muscarinic acetylcholine receptors. From there, the Ca2+ wave propagates through gap junction signaling, reaching also distant ciliated and secretory cells. These effector cells translate activation into enhanced ciliary activity and Cl- secretion, which are synergistic in boosting mucociliary clearance, the major innate defense mechanism of the airways. Our data establish tracheal brush cells as a central hub in triggering a global epithelial defense program in response to a danger-associated metabolite.
Collapse
Affiliation(s)
- Alexander Perniss
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Brett Boonen
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
- Laboratory of Ion Channel Research, VIB Center for Brain and Disease, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Moritz Thiel
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Krupali Poharkar
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Mohamad Wessam Alnouri
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Maryam Keshavarz
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Tamara Papadakis
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Silke Wiegand
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Uwe Pfeil
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Katrin Richter
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University, Giessen, Germany
| | - Mike Althaus
- Physiology Group, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Burkhard Schütz
- Institute of Anatomy and Cell Biology, Philipps University Marburg, Marburg, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Stefan Offermanns
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Trese Leinders-Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Wolfgang Kummer
- Institute of Anatomy and Cell Biology, German Center for Lung Research, Justus Liebig University Giessen; Giessen, Germany
- Excellence Cluster The Cardio-Pulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
5
|
Gsell M, Bulliard X, Schorderet Weber S, Xiang Y, Constant S, Steiner S, Biselli S, Pugin R, Palmieri M, Hogg A, Peitsch MC, Hoeng J, Stan A. Inactivation of SARS-CoV-2 on salt-coated surfaces: an in vitro study. Arch Microbiol 2023; 205:272. [PMID: 37391548 DOI: 10.1007/s00203-023-03614-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 04/28/2023] [Accepted: 06/23/2023] [Indexed: 07/02/2023]
Abstract
In the COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), face masks have become a very important safety measure against the main route of transmission of the virus: droplets and aerosols. Concerns that masks contaminated with SARS-CoV-2 infectious particles could be a risk for self-contamination have emerged early in the pandemic as well as solutions to mitigate this risk. The coating of masks with sodium chloride, an antiviral and non-hazardous to health chemical, could be an option for reusable masks. To assess the antiviral properties of salt coatings deposited onto common fabrics by spraying and dipping, the present study established an in vitro bioassay using three-dimensional airway epithelial cell cultures and SARS-CoV-2 virus. Virus particles were given directly on salt-coated material, collected, and added to the cell cultures. Infectious virus particles were measured by plaque forming unit assay and in parallel viral genome copies were quantified over time. Relative to noncoated material, the sodium chloride coating significantly reduced virus replication, confirming the effectiveness of the method to prevent fomite contamination with SARS-CoV-2. In addition, the lung epithelia bioassay proved to be suitable for future evaluation of novel antiviral coatings.
Collapse
Affiliation(s)
- Monika Gsell
- Institute for Infectious Diseases, University of Berne, 3010, Bern, Switzerland.
- Swiss Institute for Translational and Entrepreneurial Medicine, 3010, Berne, Switzerland.
| | - Xavier Bulliard
- Centre Suisse d'Electronique et de Microtechnique SA (CSEM), Rue Jaquet-Droz 1, 2002, Neuchâtel, Switzerland
| | | | - Yang Xiang
- PMI R&D, Philip Morris Products S.A., Quai Jeanreaud 5, 2000, Neuchâtel, Switzerland
| | - Samuel Constant
- Epithelix Sàrl, Chemmin des Aulx 18, Plan-Les-Ouates, 1228, Geneva, Switzerland
| | - Sandro Steiner
- PMI R&D, Philip Morris Products S.A., Quai Jeanreaud 5, 2000, Neuchâtel, Switzerland
| | - Silvia Biselli
- Centre Suisse d'Electronique et de Microtechnique SA (CSEM), Rue Jaquet-Droz 1, 2002, Neuchâtel, Switzerland
| | - Raphael Pugin
- Centre Suisse d'Electronique et de Microtechnique SA (CSEM), Rue Jaquet-Droz 1, 2002, Neuchâtel, Switzerland
| | - Michele Palmieri
- Centre Suisse d'Electronique et de Microtechnique SA (CSEM), Rue Jaquet-Droz 1, 2002, Neuchâtel, Switzerland
| | - Andreas Hogg
- Coat-X SA, Eplatures-Grise 17, 2900, La Chaux-de-Fonds, Switzerland
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A., Quai Jeanreaud 5, 2000, Neuchâtel, Switzerland
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Quai Jeanreaud 5, 2000, Neuchâtel, Switzerland
| | - Adrian Stan
- PMI R&D, Philip Morris Products S.A., Quai Jeanreaud 5, 2000, Neuchâtel, Switzerland
| |
Collapse
|
6
|
Orfali R, AlFaiz A, Rahman MA, Lau L, Nam YW, Zhang M. K Ca2 and K Ca3.1 Channels in the Airways: A New Therapeutic Target. Biomedicines 2023; 11:1780. [PMID: 37509419 PMCID: PMC10376499 DOI: 10.3390/biomedicines11071780] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
K+ channels are involved in many critical functions in lung physiology. Recently, the family of Ca2+-activated K+ channels (KCa) has received more attention, and a massive amount of effort has been devoted to developing selective medications targeting these channels. Within the family of KCa channels, three small-conductance Ca2+-activated K+ (KCa2) channel subtypes, together with the intermediate-conductance KCa3.1 channel, are voltage-independent K+ channels, and they mediate Ca2+-induced membrane hyperpolarization. Many KCa2 channel members are involved in crucial roles in physiological and pathological systems throughout the body. In this article, different subtypes of KCa2 and KCa3.1 channels and their functions in respiratory diseases are discussed. Additionally, the pharmacology of the KCa2 and KCa3.1 channels and the link between these channels and respiratory ciliary regulations will be explained in more detail. In the future, specific modulators for small or intermediate Ca2+-activated K+ channels may offer a unique therapeutic opportunity to treat muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Razan Orfali
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Ali AlFaiz
- Biomedical Research Administration, Research Centre, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia
| | - Mohammad Asikur Rahman
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Liz Lau
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Young-Woo Nam
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Miao Zhang
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
7
|
Rehman T, Welsh MJ. Inflammation as a Regulator of the Airway Surface Liquid pH in Cystic Fibrosis. Cells 2023; 12:1104. [PMID: 37190013 PMCID: PMC10137218 DOI: 10.3390/cells12081104] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The airway surface liquid (ASL) is a thin sheet of fluid that covers the luminal aspect of the airway epithelium. The ASL is a site of several first-line host defenses, and its composition is a key factor that determines respiratory fitness. Specifically, the acid-base balance of ASL has a major influence on the vital respiratory defense processes of mucociliary clearance and antimicrobial peptide activity against inhaled pathogens. In the inherited disorder cystic fibrosis (CF), loss of cystic fibrosis transmembrane conductance regulator (CFTR) anion channel function reduces HCO3- secretion, lowers the pH of ASL (pHASL), and impairs host defenses. These abnormalities initiate a pathologic process whose hallmarks are chronic infection, inflammation, mucus obstruction, and bronchiectasis. Inflammation is particularly relevant as it develops early in CF and persists despite highly effective CFTR modulator therapy. Recent studies show that inflammation may alter HCO3- and H+ secretion across the airway epithelia and thus regulate pHASL. Moreover, inflammation may enhance the restoration of CFTR channel function in CF epithelia exposed to clinically approved modulators. This review focuses on the complex relationships between acid-base secretion, airway inflammation, pHASL regulation, and therapeutic responses to CFTR modulators. These factors have important implications for defining optimal ways of tackling CF airway inflammation in the post-modulator era.
Collapse
Affiliation(s)
- Tayyab Rehman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael J. Welsh
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
8
|
Canella R, Benedusi M, Vallese A, Pecorelli A, Guiotto A, Ferrara F, Rispoli G, Cervellati F, Valacchi G. The role of potassium current in the pulmonary response to environmental oxidative stress. Arch Biochem Biophys 2023; 737:109534. [PMID: 36740034 DOI: 10.1016/j.abb.2023.109534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/30/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Exposure of human lung epithelial cells (A549 cell line) to the oxidant pollutant ozone (O3) alters cell membrane currents inducing its decrease, when the cell undergoes to a voltage-clamp protocol ranging from -90 to +70mV. The membrane potential of these cells is mainly maintained by the interplay of potassium and chloride currents. Our previous studies indicated the ability of O3 to activate ORCC (Outward Rectifier Chloride Channel) and consequently increases the chloride current. In this paper our aim was to understand the response of potassium current to oxidative stress challenge and to identify the kind potassium channel involved in O3 induced current changes. After measuring the total membrane current using an intracellular solution with or without potassium ions, we obtained the contribution of potassium to the overall membrane current in control condition by a mathematical approach. Repeating these experiments after O3 treatment we observed a significant decrease of Ipotassium. Treatment of the cells with Iberiotoxin (IbTx), a specific inhibitor of BK channel, we were able to verify the presence and the functionality of BK channels. In addition, the administration of 4-Aminopyridine (an inhibitor of voltage dependent K channels but not BK channels) and Tetraethylammonium (TEA) before and after O3 treatment we observed the formation of BK oxidative post-translation modifications. Our data suggest that O3 is able to inhibit potassium current by targeting BK channel. Further studies are needed to better clarify the role of this BK channel and its interplay with the other membrane channels under oxidative stress conditions. These findings can contribute to identify the biomolecular pathway induced by O3 allowing a possible pharmacological intervention against oxidative stress damage in lung tissue.
Collapse
Affiliation(s)
- Rita Canella
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy.
| | - Mascia Benedusi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Andrea Vallese
- Department of Environmental Sciences and Prevention, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Alessandra Pecorelli
- Department of Environmental Sciences and Prevention, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Anna Guiotto
- Department of Environmental Sciences and Prevention, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Francesca Ferrara
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Giorgio Rispoli
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Franco Cervellati
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Environmental Sciences and Prevention, University of Ferrara, Via L. Borsari, 46, Ferrara, Italy; NC State University, Plants for Human Health Institute, Animal Science Dept. NC Research Campus 600 Laureate Way, Kannapolis, NC, 28081, USA; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
9
|
Ghodake V, Dhoble S, Vavilala SL, Patravale V. Anti-biofilm potential against P. aeruginosa biofilm in cystic fibrosis infection by systemically developed garlic extract incorporated liposomal formulation. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
10
|
Baines DL, Vasiljevs S, Kalsi KK. Getting sweeter: new evidence for glucose transporters in specific cell types of the airway? Am J Physiol Cell Physiol 2023; 324:C153-C166. [PMID: 36409177 PMCID: PMC9829484 DOI: 10.1152/ajpcell.00140.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
New technologies such as single-cell RNA sequencing (scRNAseq) has enabled identification of the mRNA transcripts expressed by individual cells. This review provides insight from recent scRNAseq studies on the expression of glucose transporters in the epithelial cells of the airway epithelium from trachea to alveolus. The number of studies analyzed was limited, not all reported the full range of glucose transporters and there were differences between cells freshly isolated from the airways and those grown in vitro. Furthermore, glucose transporter mRNA transcripts were expressed at lower levels than other epithelial marker genes. Nevertheless, these studies highlighted that there were differences in cellular expression of glucose transporters. GLUT1 was the most abundant of the broadly expressed transporters that included GLUT8, 10, and 13. GLUT9 transcripts were more common in basal cells and GLUT12 in ionocytes/ciliated cells. In addition to alveolar cells, SGLT1 transcripts were present in secretory cells. GLUT3 mRNA transcripts were expressed in a cell cluster that expressed monocarboxylate (MCT2) transporters. Such distributions likely underlie cell-specific metabolic requirements to support proliferation, ion transport, mucous secretion, environment sensing, and airway glucose homeostasis. These studies have also highlighted the role of glucose transporters in the movement of dehydroascorbic acid/vitamin C/myoinositol/urate, which are factors important to the innate immune properties of the airways. Discrepancies remain between detection of mRNAs, protein, and function of glucose transporters in the lungs. However, collation of the data from further scRNAseq studies may provide a better consensus and understanding, supported by qPCR, immunohistochemistry, and functional experiments.
Collapse
Affiliation(s)
- Deborah L. Baines
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Stanislavs Vasiljevs
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| | - Kameljit K. Kalsi
- Institute for Infection and Immunity, St George’s, University of London, London, United Kingdom
| |
Collapse
|
11
|
Qiu ZE, Chen L, Hou XC, Sheng J, Xu JB, Xu JW, Gao DD, Huang ZX, Lei TL, Huang ZY, Peng L, Yang HL, Lin QH, Zhu YX, Guan WJ, Lun ZR, Zhou WL, Zhang YL. Toxoplasma gondii infection triggers ongoing inflammation mediated by increased intracellular Cl - concentration in airway epithelium. J Infect 2023; 86:47-59. [PMID: 36334726 DOI: 10.1016/j.jinf.2022.10.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 10/03/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022]
Abstract
Toxoplasma gondii is a widespread parasitic protozoan causing toxoplasmosis including pulmonary toxoplasmosis. As the first line of host defense, airway epithelial cells play critical roles in orchestrating pulmonary innate immunity. However, the mechanism underlying the airway inflammation induced by the T. gondii infection remains largely unclear. This study demonstrated that after infection with T. gondii, the major anion channel located in the apical membranes of airway epithelial cells, cystic fibrosis transmembrane conductance regulator (CFTR), was degraded by the parasite-secreted cysteine proteases. The intracellular Cl- concentration ([Cl-]i) was consequently elevated, leading to activation of nuclear factor-κB (NF-κB) signaling via serum/glucocorticoid regulated kinase 1. Furthermore, the heightened [Cl-]i and activated NF-κB signaling could be sustained in a positive feedback regulatory manner resulting from decreased intracellular cAMP level through NF-κB-mediated up-regulation of phosphodiesterase 4. Conversely, the sulfur-containing compound allicin conferred anti-inflammatory effects on pulmonary toxoplasmosis by decreasing [Cl-]i via activation of CFTR. These results suggest that the intracellular Cl- dynamically modulated by T. gondii mediates sustained airway inflammation, which provides a potential therapeutic target against pulmonary toxoplasmosis.
Collapse
Affiliation(s)
- Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jie Sheng
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jian-Bang Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Jia-Wen Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Dong-Dong Gao
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P. R. China
| | - Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Tian-Lun Lei
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Zi-Yang Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Lei Peng
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Hai-Long Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Qin-Hua Lin
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Zhao-Rong Lun
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China.
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China.
| |
Collapse
|
12
|
Mitropoulou G, Balmpouzis Z, Plojoux J, Dotta-Celio J, Sauty A, Koutsokera A. Effects of elexacaftor–tezacaftor–ivacaftor discontinuation in cystic fibrosis. Respir Med Res 2022; 82:100972. [DOI: 10.1016/j.resmer.2022.100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/12/2022]
|
13
|
Rodenburg LW, Delpiano L, Railean V, Centeio R, Pinto MC, Smits SMA, van der Windt IS, van Hugten CFJ, van Beuningen SFB, Rodenburg RNP, van der Ent CK, Amaral MD, Kunzelmann K, Gray MA, Beekman JM, Amatngalim GD. Drug Repurposing for Cystic Fibrosis: Identification of Drugs That Induce CFTR-Independent Fluid Secretion in Nasal Organoids. Int J Mol Sci 2022; 23:12657. [PMID: 36293514 PMCID: PMC9603984 DOI: 10.3390/ijms232012657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
Abstract
Individuals with cystic fibrosis (CF) suffer from severe respiratory disease due to a genetic defect in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which impairs airway epithelial ion and fluid secretion. New CFTR modulators that restore mutant CFTR function have been recently approved for a large group of people with CF (pwCF), but ~19% of pwCF cannot benefit from CFTR modulators Restoration of epithelial fluid secretion through non-CFTR pathways might be an effective treatment for all pwCF. Here, we developed a medium-throughput 384-well screening assay using nasal CF airway epithelial organoids, with the aim to repurpose FDA-approved drugs as modulators of non-CFTR-dependent epithelial fluid secretion. From a ~1400 FDA-approved drug library, we identified and validated 12 FDA-approved drugs that induced CFTR-independent fluid secretion. Among the hits were several cAMP-mediating drugs, including β2-adrenergic agonists. The hits displayed no effects on chloride conductance measured in the Ussing chamber, and fluid secretion was not affected by TMEM16A, as demonstrated by knockout (KO) experiments in primary nasal epithelial cells. Altogether, our results demonstrate the use of primary nasal airway cells for medium-scale drug screening, target validation with a highly efficient protocol for generating CRISPR-Cas9 KO cells and identification of compounds which induce fluid secretion in a CFTR- and TMEM16A-indepent manner.
Collapse
Affiliation(s)
- Lisa W. Rodenburg
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Livia Delpiano
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Violeta Railean
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| | - Raquel Centeio
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany
| | - Madalena C. Pinto
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| | - Shannon M. A. Smits
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Isabelle S. van der Windt
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Casper F. J. van Hugten
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Sam F. B. van Beuningen
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB Utrecht, The Netherlands
| | - Remco N. P. Rodenburg
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Cornelis K. van der Ent
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
| | - Margarida D. Amaral
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, 1749-016 Lisboa, Portugal
| | - Karl Kunzelmann
- Physiological Institute, University of Regensburg, D-93053 Regensburg, Germany
| | - Michael A. Gray
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jeffrey M. Beekman
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584 CB Utrecht, The Netherlands
| | - Gimano D. Amatngalim
- Department of Pediatric Pulmonology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Member of ERN-LUNG, 3584 EA Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
14
|
Hollenhorst MI, Kumar P, Zimmer M, Salah A, Maxeiner S, Elhawy MI, Evers SB, Flockerzi V, Gudermann T, Chubanov V, Boehm U, Krasteva-Christ G. Taste Receptor Activation in Tracheal Brush Cells by Denatonium Modulates ENaC Channels via Ca2+, cAMP and ACh. Cells 2022; 11:cells11152411. [PMID: 35954259 PMCID: PMC9367940 DOI: 10.3390/cells11152411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/23/2022] [Accepted: 08/03/2022] [Indexed: 02/04/2023] Open
Abstract
Mucociliary clearance is a primary defence mechanism of the airways consisting of two components, ciliary beating and transepithelial ion transport (ISC). Specialised chemosensory cholinergic epithelial cells, named brush cells (BC), are involved in regulating various physiological and immunological processes. However, it remains unclear if BC influence ISC. In murine tracheae, denatonium, a taste receptor agonist, reduced basal ISC in a concentration-dependent manner (EC50 397 µM). The inhibition of bitter taste signalling components with gallein (Gβγ subunits), U73122 (phospholipase C), 2-APB (IP3-receptors) or with TPPO (Trpm5, transient receptor potential-melastatin 5 channel) reduced the denatonium effect. Supportively, the ISC was also diminished in Trpm5−/− mice. Mecamylamine (nicotinic acetylcholine receptor, nAChR, inhibitor) and amiloride (epithelial sodium channel, ENaC, antagonist) decreased the denatonium effect. Additionally, the inhibition of Gα subunits (pertussis toxin) reduced the denatonium effect, while an inhibition of phosphodiesterase (IBMX) increased and of adenylate cyclase (forskolin) reversed the denatonium effect. The cystic fibrosis transmembrane conductance regulator (CFTR) inhibitor CFTRinh172 and the KCNQ1 potassium channel antagonist chromanol 293B both reduced the denatonium effect. Thus, denatonium reduces ISC via the canonical bitter taste signalling cascade leading to the Trpm5-dependent nAChR-mediated inhibition of ENaC as well as Gα signalling leading to a reduction in cAMP-dependent ISC. Therefore, BC activation contributes to the regulation of fluid homeostasis.
Collapse
Affiliation(s)
| | - Praveen Kumar
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Maxim Zimmer
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Alaa Salah
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | | | - Saskia B. Evers
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
| | - Veit Flockerzi
- Institute for Experimental and Clinical Pharmacology and Toxicology, Centre for Molecular Signalling, Saarland University, 66421 Homburg, Germany
| | - Thomas Gudermann
- Walter-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University and German Centre for Lung Research (DZL), 80366 Munich, Germany
| | - Vladimir Chubanov
- Walter-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University and German Centre for Lung Research (DZL), 80366 Munich, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Centre for Molecular Signalling, School of Medicine, Saarland University, 66421 Homburg, Germany
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg, Germany
- Correspondence: ; Tel.: +49-6841-16-26101
| |
Collapse
|
15
|
Zhou W, Yu T, Hua Y, Hou Y, Ding Y, Nie H. Effects of Hypoxia on Respiratory Diseases: Perspective View of Epithelial Ion Transport. Am J Physiol Lung Cell Mol Physiol 2022; 323:L240-L250. [PMID: 35819839 DOI: 10.1152/ajplung.00065.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The balance of gas exchange and lung ventilation is essential for the maintenance of body homeostasis. There are many ion channels and transporters in respiratory epithelial cells, including epithelial sodium channel, Na,K-ATPase, cystic fibrosis transmembrane conductance regulator, and some transporters. These ion channels/transporters maintain the capacity of liquid layer on the surface of respiratory epithelial cells, and provide an immune barrier for the respiratory system to clear off foreign pathogens. However, in some harmful external environment and/or pathological conditions, the respiratory epithelium is prone to hypoxia, which would destroy the ion transport function of the epithelium and unbalance the homeostasis of internal environment, triggering a series of pathological reactions. Many respiratory diseases associated with hypoxia manifest an increased expression of hypoxia-inducible factor-1, which mediates the integrity of the epithelial barrier and affects epithelial ion transport function. It is important to study the relationship between hypoxia and ion transport function, whereas the mechanism of hypoxia-induced ion transport dysfunction in respiratory diseases is not clear. This review focuses on the relationship of hypoxia and respiratory diseases, as well as dysfunction of ion transport and tight junctions in respiratory epithelial cells under hypoxia.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Hua
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
16
|
Brown EF, Mitaera T, Fronius M. COVID-19 and Liquid Homeostasis in the Lung—A Perspective through the Epithelial Sodium Channel (ENaC) Lens. Cells 2022; 11:cells11111801. [PMID: 35681496 PMCID: PMC9180030 DOI: 10.3390/cells11111801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 01/26/2023] Open
Abstract
Infections with a new corona virus in 2019 lead to the definition of a new disease known as Corona Virus Disease 2019 (COVID-19). The sever cases of COVID-19 and the main cause of death due to virus infection are attributed to respiratory distress. This is associated with the formation of pulmonary oedema that impairs blood oxygenation and hypoxemia as main symptoms of respiratory distress. An important player for the maintenance of a defined liquid environment in lungs needed for normal lung function is the epithelial sodium channel (ENaC). The present article reviews the implications of SARS-CoV-2 infections from the perspective of impaired function of ENaC. The rationale for this perspective is derived from the recognition that viral spike protein and ENaC share a common proteolytic cleavage site. This cleavage site is utilized by the protease furin, that is essential for ENaC activity. Furin cleavage of spike ‘activates’ the virus protein to enable binding to host cell membrane receptors and initiate cell infection. Based on the importance of proteolytic cleavage for ENaC function and activation of spike, it seems feasible to assume that virus infections are associated with impaired ENaC activity. This is further supported by symptoms of COVID-19 that are reminiscent of impaired ENaC function in the respiratory tract.
Collapse
Affiliation(s)
- Emily F. Brown
- Department of Physiology, University of Otago, Dunedin 9054, New Zealand; (E.F.B.); (T.M.)
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Tamapuretu Mitaera
- Department of Physiology, University of Otago, Dunedin 9054, New Zealand; (E.F.B.); (T.M.)
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin 9054, New Zealand; (E.F.B.); (T.M.)
- HeartOtago, University of Otago, Dunedin, New Zealand
- Healthy Hearts for Aotearoa New Zealand, Centre of Research Excellence, New Zealand
- Maurice Wilkins Centre for Molecular Discovery, Centre of Research Excellence, New Zealand
- Correspondence: ; Tel.: +64-3-471-6081
| |
Collapse
|
17
|
Luettich K, Sharma M, Yepiskoposyan H, Breheny D, Lowe FJ. An Adverse Outcome Pathway for Decreased Lung Function Focusing on Mechanisms of Impaired Mucociliary Clearance Following Inhalation Exposure. FRONTIERS IN TOXICOLOGY 2022; 3:750254. [PMID: 35295103 PMCID: PMC8915806 DOI: 10.3389/ftox.2021.750254] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/11/2021] [Indexed: 01/23/2023] Open
Abstract
Adverse outcome pathways (AOPs) help to organize available mechanistic information related to an adverse outcome into key events (KEs) spanning all organizational levels of a biological system(s). AOPs, therefore, aid in the biological understanding of a particular pathogenesis and also help with linking exposures to eventual toxic effects. In the regulatory context, knowledge of disease mechanisms can help design testing strategies using in vitro methods that can measure or predict KEs relevant to the biological effect of interest. The AOP described here evaluates the major processes known to be involved in regulating efficient mucociliary clearance (MCC) following exposures causing oxidative stress. MCC is a key aspect of the innate immune defense against airborne pathogens and inhaled chemicals and is governed by the concerted action of its functional components, the cilia and airway surface liquid (ASL). The AOP network described here consists of sequences of KEs that culminate in the modulation of ciliary beat frequency and ASL height as well as mucus viscosity and hence, impairment of MCC, which in turn leads to decreased lung function.
Collapse
Affiliation(s)
- Karsta Luettich
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Monita Sharma
- PETA Science Consortium International e.V., Stuttgart, Germany
| | - Hasmik Yepiskoposyan
- Philip Morris International R&D, Philip Morris Products S.A., Neuchatel, Switzerland
| | - Damien Breheny
- British American Tobacco (Investments) Ltd., Group Research and Development, Southampton, United Kingdom
| | - Frazer J Lowe
- Broughton Nicotine Services, Earby, Lancashire, United Kingdom
| |
Collapse
|
18
|
Simões FB, Kmit A, Amaral MD. Cross-talk of inflammatory mediators and airway epithelium reveals the cystic fibrosis transmembrane conductance regulator as a major target. ERJ Open Res 2021; 7:00247-2021. [PMID: 34912883 PMCID: PMC8666577 DOI: 10.1183/23120541.00247-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/13/2021] [Indexed: 11/05/2022] Open
Abstract
Airway inflammation, mucus hyperproduction and epithelial remodelling are hallmarks of many chronic airway diseases, including asthma, COPD and cystic fibrosis. While several cytokines are dysregulated in these diseases, most studies focus on the response of airways to interleukin (IL)-4 and IL-13, which have been shown to induce mucus hyperproduction and shift the airway epithelium towards a hypersecretory phenotype. We hypothesised that other cytokines might induce the expression of chloride (Cl-) channels/transporters, and regulate epithelial differentiation and mucus production. To this end, fully differentiated human airway basal cells (BCi-NS1.1) were treated with cytokines identified as dysregulated in those diseases, namely IL-8, IL-1β, IL-4, IL-17A, IL-10 and IL-22, and tumour necrosis factor-α. Our results show that the cystic fibrosis transmembrane conductance regulator (CFTR) is the main Cl- channel modulated by inflammation, in contrast to transmembrane protein 16A (TMEM16A), whose levels only changed with IL-4. Furthermore, we identified novel roles for IL-10 and IL-22 by influencing epithelial differentiation towards ciliated cells and away from pulmonary ionocytes. In contrast, IL-1β and IL-4 reduced the number of ciliated cells while increasing club cells. Interestingly, while IL-1β, IL-4 and IL-10 upregulated CFTR expression, IL-4 was the only cytokine that increased both its function and the number of CFTR-expressing club cells, suggesting that this cell type may be the main contributor for CFTR function. Additionally, all cytokines assessed increased mucus production through a differential upregulation of MUC5AC and MUC5B transcript levels. This study reveals a novel insight into differentiation resulting from the cross-talk of inflammatory mediators and airway epithelial cells, which is particularly relevant for chronic airway diseases.
Collapse
Affiliation(s)
- Filipa B Simões
- Faculty of Sciences, University of Lisbon, BioISI - Biosystems and Integrative Sciences Institute, Lisbon, Portugal
| | - Arthur Kmit
- Faculty of Sciences, University of Lisbon, BioISI - Biosystems and Integrative Sciences Institute, Lisbon, Portugal
| | - Margarida D Amaral
- Faculty of Sciences, University of Lisbon, BioISI - Biosystems and Integrative Sciences Institute, Lisbon, Portugal
| |
Collapse
|
19
|
Lagowala DA, Kwon S, Sidhaye VK, Kim DH. Human microphysiological models of airway and alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1072-L1088. [PMID: 34612064 PMCID: PMC8715018 DOI: 10.1152/ajplung.00103.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/22/2022] Open
Abstract
Human organ-on-a-chip models are powerful tools for preclinical research that can be used to study the mechanisms of disease and evaluate new targets for therapeutic intervention. Lung-on-a-chip models have been one of the most well-characterized designs in this field and can be altered to evaluate various types of respiratory disease and to assess treatment candidates prior to clinical testing. These systems are capable of overcoming the flaws of conventional two-dimensional (2-D) cell culture and in vivo animal testing due to their ability to accurately recapitulate the in vivo microenvironment of human tissue with tunable material properties, microfluidic integration, delivery of precise mechanical and biochemical cues, and designs with organ-specific architecture. In this review, we first describe an overview of currently available lung-on-a-chip designs. We then present how recent innovations in human stem cell biology, tissue engineering, and microfabrication can be used to create more predictive human lung-on-a-chip models for studying respiratory disease. Finally, we discuss the current challenges and future directions of lung-on-a-chip designs for in vitro disease modeling with a particular focus on immune and multiorgan interactions.
Collapse
Affiliation(s)
- Dave Anuj Lagowala
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Seoyoung Kwon
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Venkataramana K Sidhaye
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
Brookes O, Boland S, Lai Kuen R, Miremont D, Movassat J, Baeza-Squiban A. Co-culture of type I and type II pneumocytes as a model of alveolar epithelium. PLoS One 2021; 16:e0248798. [PMID: 34570783 PMCID: PMC8475999 DOI: 10.1371/journal.pone.0248798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/13/2021] [Indexed: 11/18/2022] Open
Abstract
The epithelial tissues of the distal lung are continuously exposed to inhaled air, and are of research interest in studying respiratory exposure to both hazardous and therapeutic materials. Pharmaco-toxicological research depends on the development of sophisticated models of the alveolar epithelium, which better represent the different cell types present in the native lung and interactions between them. We developed an air-liquid interface (ALI) model of the alveolar epithelium which incorporates cell lines which bear features of type I (hAELVi) and type II (NCI-H441) epithelial cells. We compared morphology of single cells and the structure of cell layers of the two lines using light and electron microscopy. Working both in monotypic cultures and cocultures, we measured barrier function by trans-epithelial electrical resistance (TEER), and demonstrated that barrier properties can be maintained for 30 days. We created a mathematical model of TEER development over time based on these data in order to make inferences about the interactions occurring in these culture systems. We assessed expression of a panel of relevant genes that play important roles in barrier function and differentiation. The coculture model was observed to form a stable barrier akin to that seen in hAELVi, while expressing surfactant protein C, and having a profile of expression of claudins and aquaporins appropriate for the distal lung. We described cavities which arise within stratified cell layers in NCI-H441 and cocultured cells, and present evidence that these cavities represent an aberrant apical surface. In summary, our results support the coculture of these two cell lines to produce a model which better represents the breadth of functions seen in native alveolar epithelium.
Collapse
Affiliation(s)
- Oliver Brookes
- Unité de Biologie Fonctionnelle et Adaptative UMR 8251, CNRS, Université de Paris, Paris, France
| | - Sonja Boland
- Unité de Biologie Fonctionnelle et Adaptative UMR 8251, CNRS, Université de Paris, Paris, France
| | - René Lai Kuen
- Cellular and Molecular Imaging Facility, US25 Inserm—3612 CNRS, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Dorian Miremont
- Unité de Biologie Fonctionnelle et Adaptative UMR 8251, CNRS, Université de Paris, Paris, France
| | - Jamileh Movassat
- Unité de Biologie Fonctionnelle et Adaptative UMR 8251, CNRS, Université de Paris, Paris, France
| | - Armelle Baeza-Squiban
- Unité de Biologie Fonctionnelle et Adaptative UMR 8251, CNRS, Université de Paris, Paris, France
- * E-mail:
| |
Collapse
|
21
|
Schmid B, Kredel M, Ullrich R, Krenn K, Lucas R, Markstaller K, Fischer B, Kranke P, Meybohm P, Zwißler B, Frank S. Safety and preliminary efficacy of sequential multiple ascending doses of solnatide to treat pulmonary permeability edema in patients with moderate-to-severe ARDS-a randomized, placebo-controlled, double-blind trial. Trials 2021; 22:643. [PMID: 34544463 PMCID: PMC8450703 DOI: 10.1186/s13063-021-05588-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a complex clinical diagnosis with various possible etiologies. One common feature, however, is pulmonary permeability edema, which leads to an increased alveolar diffusion pathway and, subsequently, impaired oxygenation and decarboxylation. A novel inhaled peptide agent (AP301, solnatide) was shown to markedly reduce pulmonary edema in animal models of ARDS and to be safe to administer to healthy humans in a Phase I clinical trial. Here, we present the protocol for a Phase IIB clinical trial investigating the safety and possible future efficacy endpoints in ARDS patients. Methods This is a randomized, placebo-controlled, double-blind intervention study. Patients with moderate to severe ARDS in need of mechanical ventilation will be randomized to parallel groups receiving escalating doses of solnatide or placebo, respectively. Before advancing to a higher dose, a data safety monitoring board will investigate the data from previous patients for any indication of patient safety violations. The intervention (application of the investigational drug) takes places twice daily over the course of 7 days, ensued by a follow-up period of another 21 days. Discussion The patients to be included in this trial will be severely sick and in need of mechanical ventilation. The amount of data to be collected upon screening and during the course of the intervention phase is substantial and the potential timeframe for inclusion of any given patient is short. However, when prepared properly, adherence to this protocol will make for the acquisition of reliable data. Particular diligence needs to be exercised with respect to informed consent, because eligible patients will most likely be comatose and/or deeply sedated at the time of inclusion. Trial registration This trial was prospectively registered with the EU Clinical trials register (clinicaltrialsregister.eu). EudraCT Number: 2017-003855-47.
Collapse
Affiliation(s)
- Benedikt Schmid
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Markus Kredel
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Roman Ullrich
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Katharina Krenn
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Rudolf Lucas
- Vascular Biology Center, Division of Pulmonary Medicine, Medical College of Georgia, Augusta University, Augusta, USA
| | - Klaus Markstaller
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Peter Kranke
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany.
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Bernhard Zwißler
- Department of Anesthesiology, University Hospital of Ludwig-Maximilians-University (LMU), Munich, Germany.,Comprehensive Pulmonary Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Sandra Frank
- Department of Anesthesiology, University Hospital of Ludwig-Maximilians-University (LMU), Munich, Germany
| | | |
Collapse
|
22
|
Huijghebaert S, Hoste L, Vanham G. Essentials in saline pharmacology for nasal or respiratory hygiene in times of COVID-19. Eur J Clin Pharmacol 2021; 77:1275-1293. [PMID: 33772626 PMCID: PMC7998085 DOI: 10.1007/s00228-021-03102-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/28/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Nasal irrigation or nebulizing aerosol of isotonic or hypertonic saline is a traditional method for respiratory or nasal care. A recent small study in outpatients with COVID-19 without acute respiratory distress syndrome suggests substantial symptom resolution. We therefore analyzed pharmacological/pharmacodynamic effects of isotonic or hypertonic saline, relevant to SARS-CoV-2 infection and respiratory care. METHODS Mixed search method. RESULTS Due to its wetting properties, saline achieves an improved spreading of alveolar lining fluid and has been shown to reduce bio-aerosols and viral load. Saline provides moisture to respiratory epithelia and gels mucus, promotes ciliary beating, and improves mucociliary clearance. Coronaviruses and SARS-CoV-2 damage ciliated epithelium in the nose and airways. Saline inhibits SARS-CoV-2 replication in Vero cells; possible interactions involve the viral ACE2-entry mechanism (chloride-dependent ACE2 configuration), furin and 3CLpro (inhibition by NaCl), and the sodium channel ENaC. Saline shifts myeloperoxidase activity in epithelial or phagocytic cells to produce hypochlorous acid. Clinically, nasal or respiratory airway care with saline reduces symptoms of seasonal coronaviruses and other common cold viruses. Its use as aerosol reduces hospitalization rates for bronchiolitis in children. Preliminary data suggest symptom reduction in symptomatic COVID-19 patients if saline is initiated within 48 h of symptom onset. CONCLUSIONS Saline interacts at various levels relevant to nasal or respiratory hygiene (nasal irrigation, gargling or aerosol). If used from the onset of common cold symptoms, it may represent a useful add-on to first-line interventions for COVID-19. Formal evaluation in mild COVID-19 is desirable as to establish efficacy and optimal treatment regimens.
Collapse
Affiliation(s)
| | - Levi Hoste
- Pediatric Pulmonology, Infectious Diseases and Immunology, Ghent University Hospital, Ghent, Belgium
- Primary Immunodeficiency Research Lab, Center for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Center, Ghent University Hospital, Ghent, Belgium
| | - Guido Vanham
- Department of Biomedical Sciences, Institute of Tropical Medicine and University of Antwerp, Antwerp, Belgium
| |
Collapse
|
23
|
Ambade V, Ambade S. “SARS-CoV-2 infection of endothelial cell, clinical laboratory and autopsy findings, and outcomes suggest role of hypoxia-inducible factor-1 in COVID-19”. J Med Biochem 2021; 41:14-20. [PMID: 35291496 PMCID: PMC8882013 DOI: 10.5937/jomb0-30659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/13/2021] [Indexed: 11/28/2022] Open
Abstract
Researchers around the world have experienced the dual nature of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), 'tragically lethal in some people while surprisingly benign in others'. There have been congregating studies of the novel coronavirus disease (COVID-19), a disease that mainly attacks the lungs but also has mystifying effects on the heart, kidneys and brain. Researchers are also gathering information to ascertain why people are dying of COVID-19, whether it is solely a respiratory disorder, a coagulation disorder or multi-organ failure. Alterations in laboratory parameters like lactate, ferritin and albumin have been established as risk factors and are associated with outcomes, yet none have not been sub stantiated with a scientific biochemical rationale. SARSCoV-2 affects the alveolar type II epithelial cells which significantly disturbs its surfactant homeostasis, deprives Na,K-ATPase of ATP, thereby disturbing the alveolar lining fluid which then gradually decreases the alveolar gaseous exchange initiating the intracellular hypoxic conditions. This activates AMP-activated kinase, which further inhibits Na,K-ATPase, which can progressively cause respiratory distress syndrome. The virus may infect endothelial cell (EC) which, being less energetic, cannot withstand the huge energy requirement towards viral replication. There - fore glycolysis, the prime energy generating pathway, must be mandatorily upregulated. This can be achieved by Hypoxia-inducible factor-1 (HIF-1). However, HIF-1 also activates transcription of von Willebrand factor, plasminogen activator inhibitor-1, and suppresses the release of thrombomodulin. This in turn sets off the coagulation cascade that can lead to in-situ pulmonary thrombosis and micro clots. The proposed HIF-1 hypothesis justifies various features, biochemical alteration, laboratory as well as autopsy findings such as respiratory distress syndrome, increased blood ferritin and lactate levels, hypoalbuminemia, endothelial invasion, in-situ pulmonary thrombosis and micro clots, and multi-organ failure in COVID-19.
Collapse
Affiliation(s)
- Vivek Ambade
- Senior Faculty, Department of Biochemistry, City Pune, State Maharashtra, India
| | - Sonia Ambade
- H V Desai College, Department of Microbiology, City Pune, State Maharashtra, India
| |
Collapse
|
24
|
Sek A, Kampa RP, Kulawiak B, Szewczyk A, Bednarczyk P. Identification of the Large-Conductance Ca 2+-Regulated Potassium Channel in Mitochondria of Human Bronchial Epithelial Cells. Molecules 2021; 26:molecules26113233. [PMID: 34072205 PMCID: PMC8199365 DOI: 10.3390/molecules26113233] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria play a key role in energy metabolism within the cell. Potassium channels such as ATP-sensitive, voltage-gated or large-conductance Ca2+-regulated channels have been described in the inner mitochondrial membrane. Several hypotheses have been proposed to describe the important roles of mitochondrial potassium channels in cell survival and death pathways. In the current study, we identified two populations of mitochondrial large-conductance Ca2+-regulated potassium (mitoBKCa) channels in human bronchial epithelial (HBE) cells. The biophysical properties of the channels were characterized using the patch-clamp technique. We observed the activity of the channel with a mean conductance close to 285 pS in symmetric 150/150 mM KCl solution. Channel activity was increased upon application of the potassium channel opener NS11021 in the micromolar concentration range. The channel activity was completely inhibited by 1 µM paxilline and 300 nM iberiotoxin, selective inhibitors of the BKCa channels. Based on calcium and iberiotoxin modulation, we suggest that the C-terminus of the protein is localized to the mitochondrial matrix. Additionally, using RT-PCR, we confirmed the presence of α pore-forming (Slo1) and auxiliary β3-β4 subunits of BKCa channel in HBE cells. Western blot analysis of cellular fractions confirmed the mitochondrial localization of α pore-forming and predominately β3 subunits. Additionally, the regulation of oxygen consumption and membrane potential of human bronchial epithelial mitochondria in the presence of the potassium channel opener NS11021 and inhibitor paxilline were also studied. In summary, for the first time, the electrophysiological and functional properties of the mitoBKCa channel in a bronchial epithelial cell line were described.
Collapse
Affiliation(s)
- Aleksandra Sek
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
- Faculty of Chemistry, University of Warsaw, 02-093 Warsaw, Poland
| | - Rafal P. Kampa
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences—SGGW, 02-776 Warsaw, Poland
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.S.); (R.P.K.); (B.K.); (A.S.)
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences—SGGW, 02-776 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-593-8620
| |
Collapse
|
25
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
26
|
Cui Y, Hou Y, Zhang H, Liu Y, Mao K, Nie H, Ding Y. Regulation of Electrolyte Permeability by Herbal Monomers in Edematous Disorders. Curr Pharm Des 2021; 27:833-839. [PMID: 32940173 DOI: 10.2174/1381612826666200917144655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
Edema is a gradual accumulation of fluid in the interstitial tissues or luminal cavities, which is regulated by ion transport pathways and reflects dysfunction of fluid and salt homeostasis. Increasing evidence suggests that some herbal monomers significantly reduce organ/tissue edema. In this review, we briefly summarized the electrolyte permeability involved in pathomechanisms of organ edema, and the benefits of herbal monomers on ionic transport machinery, including Na+-K+-ATPase, Na+ and Cl- channels, Na+-K+-2Cl- co-transporter, etc. Pharmaceutical relevance is implicated in developing advanced strategies to mitigate edematous disorders. In conclusion, the natural herbal monomers regulate electrolyte permeability in many edematous disorders, and further basic and clinical studies are needed.
Collapse
Affiliation(s)
- Yong Cui
- Department of Anesthesiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yanhong Liu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Kejun Mao
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
27
|
Role of pirfenidone in TGF-β pathways and other inflammatory pathways in acute respiratory syndrome coronavirus 2 (SARS-Cov-2) infection: a theoretical perspective. Pharmacol Rep 2021; 73:712-727. [PMID: 33880743 PMCID: PMC8057922 DOI: 10.1007/s43440-021-00255-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes pulmonary injury or multiple-organ injury by various pathological pathways. Transforming growth factor-beta (TGF-β) is a key factor that is released during SARS-CoV-2 infection. TGF-β, by internalization of the epithelial sodium channel (ENaC), suppresses the anti-oxidant system, downregulates the cystic fibrosis transmembrane conductance regulator (CFTR), and activates the plasminogen activator inhibitor 1 (PAI-1) and nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-kB). These changes cause inflammation and lung injury along with coagulopathy. Moreover, reactive oxygen species play a significant role in lung injury, which levels up during SARS-CoV-2 infection. Drug Suggestion Pirfenidone is an anti-fibrotic drug with an anti-oxidant activity that can prevent lung injury during SARS-CoV-2 infection by blocking the maturation process of transforming growth factor-beta (TGF-β) and enhancing the protective role of peroxisome proliferator-activated receptors (PPARs). Pirfenidone is a safe drug for patients with hypertension or diabetes and its side effect tolerated well. Conclusion The drug as a theoretical perspective may be an effective and safe choice for suppressing the inflammatory response during COVID-19. The recommendation would be a combination of pirfenidone and N-acetylcysteine to achieve maximum benefit during SARS-CoV-2 treatment.
Collapse
|
28
|
Munis AM, Hyde SC, Gill DR. A human surfactant B deficiency air-liquid interface cell culture model suitable for gene therapy applications. Mol Ther Methods Clin Dev 2021; 20:237-246. [PMID: 33426150 PMCID: PMC7782204 DOI: 10.1016/j.omtm.2020.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/17/2020] [Indexed: 01/02/2023]
Abstract
Surfactant protein B (SPB) deficiency is a severe monogenic interstitial lung disorder that leads to loss of life in infants as a result of alveolar collapse and respiratory distress syndrome. The development and assessment of curative therapies for the deficiency are limited by the general lack of well-characterized and physiologically relevant in vitro models of human lung parenchyma. Here, we describe a new human surfactant air-liquid interface (SALI) culture model based on H441 cells, which successfully recapitulates the key characteristics of human alveolar cells in primary culture as evidenced by RNA and protein expression of alveolar cell markers. SALI cultures were able to develop stratified cellular layers with functional barrier properties that are stable for at least 28 days after air-lift. A SFTPB knockout model of SPB deficiency was generated via gene editing of SALI cultures. The SFTPB-edited SALI cultures lost expression of SPB completely and showed weaker functional barrier properties. We were able to correct this phenotype via delivery of a lentiviral vector pseudotyped with Sendai virus glycoproteins F/HN expressing SPB. We believe that SALI cultures can serve as an important in vitro research tool to study human alveolar epithelium, especially for the development of advanced therapy medicinal products targeting monogenic disorders.
Collapse
Affiliation(s)
- Altar M. Munis
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen C. Hyde
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Deborah R. Gill
- Gene Medicine Group, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Walentek P. Xenopus epidermal and endodermal epithelia as models for mucociliary epithelial evolution, disease, and metaplasia. Genesis 2021; 59:e23406. [PMID: 33400364 DOI: 10.1002/dvg.23406] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/08/2022]
Abstract
The Xenopus embryonic epidermis is a powerful model to study mucociliary biology, development, and disease. Particularly, the Xenopus system is being used to elucidate signaling pathways, transcription factor functions, and morphogenetic mechanisms regulating cell fate specification, differentiation and cell function. Thereby, Xenopus research has provided significant insights into potential underlying molecular mechanisms for ciliopathies and chronic airway diseases. Recent studies have also established the embryonic epidermis as a model for mucociliary epithelial remodeling, multiciliated cell trans-differentiation, cilia loss, and mucus secretion. Additionally, the tadpole foregut epithelium is lined by a mucociliary epithelium, which shows remarkable features resembling mammalian airway epithelia, including its endodermal origin and a variable cell type composition along the proximal-distal axis. This review aims to summarize the advantages of the Xenopus epidermis for mucociliary epithelial biology and disease modeling. Furthermore, the potential of the foregut epithelium as novel mucociliary model system is being highlighted. Additional perspectives are presented on how to expand the range of diseases that can be modeled in the frog system, including proton pump inhibitor-associated pneumonia as well as metaplasia in epithelial cells of the airway and the gastroesophageal region.
Collapse
Affiliation(s)
- Peter Walentek
- Renal Division, Department of Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany.,CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Kamili C, Kandoti HS, Radhakrishnan S, Konde A, Vattikutti UMR. Anti-angiogenic activity of chloride and potassium channel modulators: repurposing ion channel modulators. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Excessive angiogenesis can be the root cause of many pathological conditions. Various types of ion channels are found on the endothelial cells. These ion channels play a vital role in the multi-stepped process of angiogenesis. The study aims to investigate the anti-angiogenic effects of specific ion channel modulators mefloquine (volume-regulated chloride channel blocker), lubiprostone (ClC-2 channel agonist), and 4-aminopyridine (voltage-gated potassium channel blocker).
Results
The anti-angiogenic activity of ion channel modulators was screened by measuring its effects on the area of neovascularization and histopathological studies by in vivo (corneal neovascularization) method and by in vitro assays, endothelial cell proliferation assay, cell migration assay, and matrigel cord-like morphogenesis assay. The test and standard drug (bevacizumab) groups were compared with the control group using one-way ANOVA, followed by post hoc test, and Dunnett’s test to compare the mean of all the groups with the control mean. The results revealed that mefloquine at the dose of 0.6% w/v and 1.0% w/v, lubiprostone at the dose of 0.5% w/v and 1.0% w/v, and 4-aminopyridine at the dose of 2% w/v and 4% w/v showed significant anti-angiogenic property. In the studies on human umbilical vein endothelial cells, the test drugs (100 nM) showed significant inhibition of proliferation, migration, and decrease in network length of cord-like tubes.
Conclusion
The scientific findings indicate that the test drugs have potent anti-angiogenic activity by inhibiting the cell proliferation, inhibiting the cell volume increase, arresting the cell cycle progression and by causing membrane hyperpolarization. The potent anti-angiogenic drugs obtained by repurposing these ion channel modulators, in the further studies, will be able to treat the diseases due to excess angiogenesis from the root cause.
Graphical abstract
Collapse
|
31
|
Kumar P, Scholze P, Fronius M, Krasteva-Christ G, Hollenhorst MI. Nicotine stimulates ion transport via metabotropic β4 subunit containing nicotinic ACh receptors. Br J Pharmacol 2020; 177:5595-5608. [PMID: 32959891 PMCID: PMC7707097 DOI: 10.1111/bph.15270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/25/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Mucociliary clearance is an innate immune process of the airways, essential for removal of respiratory pathogens. It depends on ciliary beat and ion and fluid homeostasis of the epithelium. We have shown that nicotinic ACh receptors (nAChRs) activate ion transport in mouse tracheal epithelium. Yet the receptor subtypes and signalling pathways involved remained unknown. Experimental Approach Transepithelial short circuit currents (ISC) of freshly isolated mouse tracheae were recorded using the Ussing chamber technique. Changes in [Ca2+]i were studied on freshly dissociated mouse tracheal epithelial cells. Key Results Apical application of the nAChR agonist nicotine transiently increased ISC. The nicotine effect was abolished by the nAChR antagonist mecamylamine. α‐Bungarotoxin (α7 antagonist) had no effect. The agonists epibatidine (α3β2, α4β2, α4β4 and α3β4) and A‐85380 (α4β2 and α3β4) increased ISC. The antagonists dihydro‐β‐erythroidine (α4β2, α3β2, α4β4 and α3β4), α‐conotoxin MII (α3β2) and α‐conotoxin PnIA (α3β2) reduced the nicotine effect. Nicotine‐ and epibatidine‐induced currents were unaltered in β2−/−mice, but in β4−/− mice no increase was observed. In the presence of thapsigargin (endoplasmatic reticulum Ca2+‐ATPase inhibitor) or the ryanodine receptor antagonists JTV‐519 and dantrolene there was a reduction in the nicotine‐effect, indicating involvement of Ca2+ release from intracellular stores. Additionally, the PKA inhibitor H‐89 and the TMEM16A (Ca2+‐activated chloride channel) inhibitor T16Ainh‐A01 significantly reduced the nicotine‐effect. Conclusion and Implications α3β4 nAChRs are responsible for the nicotine‐induced current changes via Ca2+ release from intracellular stores, PKA and ryanodine receptor activation. These nAChRs might be possible targets to stimulate chloride transport via TMEM16A.
Collapse
Affiliation(s)
- Praveen Kumar
- Institute of Anatomy and Cell Biology, Saarland University, Homburg, Germany
| | - Petra Scholze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Martin Fronius
- Department of Physiology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
32
|
Weber J, Rajan S, Schremmer C, Chao YK, Krasteva-Christ G, Kannler M, Yildirim AÖ, Brosien M, Schredelseker J, Weissmann N, Grimm C, Gudermann T, Dietrich A. TRPV4 channels are essential for alveolar epithelial barrier function as protection from lung edema. JCI Insight 2020; 5:134464. [PMID: 32931478 PMCID: PMC7605532 DOI: 10.1172/jci.insight.134464] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
Ischemia/reperfusion-induced edema (IRE), one of the most significant causes of mortality after lung transplantation, can be mimicked ex vivo in isolated perfused mouse lungs (IPL). Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel studied in endothelium; however, its role in the lung epithelium remains elusive. Here, we show enhanced IRE in TRPV4-deficient (TRPV4–/–) IPL compared with that of WT controls, indicating a protective role of TRPV4 in maintenance of the alveolar epithelial barrier. By immunohistochemistry, mRNA profiling, and electrophysiological characterization, we detected TRPV4 in bronchial epithelium, alveolar epithelial type I (ATI), and alveolar epithelial type II (ATII) cells. Genetic ablation of TRPV4 resulted in reduced expression of the water-conducting aquaporin-5 (AQP-5) channel in ATI cells. Migration of TRPV4–/– ATI cells was reduced, and cell barrier function was impaired. Analysis of isolated primary TRPV4–/– ATII cells revealed a reduced expression of surfactant protein C, and the TRPV4 activator GSK1016790A induced increases in current densities only in WT ATII cells. Moreover, TRPV4–/– lungs of adult mice developed significantly larger mean chord lengths and altered lung function compared with WT lungs. Therefore, our data illustrate essential functions of TRPV4 channels in alveolar epithelial cells and in protection from edema formation. TRPV4, a non-selective cation channel, is essential for alveolar epithelial function and protects from ischemia-reperfusion-induced lung edema.
Collapse
Affiliation(s)
- Jonas Weber
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Suhasini Rajan
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Christian Schremmer
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Yu-Kai Chao
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Gabriela Krasteva-Christ
- Institute of Anatomy and Cell Biology, School of Medicine, Saarland University, Homburg, Germany
| | - Martina Kannler
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, a member of the DZL, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Monika Brosien
- Justus Liebig University Giessen, Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center, a member of the DZL, Giessen, Germany
| | - Johann Schredelseker
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Norbert Weissmann
- Justus Liebig University Giessen, Cardio-Pulmonary Institute, University of Giessen and Marburg Lung Center, a member of the DZL, Giessen, Germany
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, a member of the German Center for Lung Research (DZL), Ludwig Maximilian University of Munich, Munich Germany
| |
Collapse
|
33
|
Scudieri P, Musante I, Venturini A, Guidone D, Genovese M, Cresta F, Caci E, Palleschi A, Poeta M, Santamaria F, Ciciriello F, Lucidi V, Galietta LJV. Ionocytes and CFTR Chloride Channel Expression in Normal and Cystic Fibrosis Nasal and Bronchial Epithelial Cells. Cells 2020; 9:cells9092090. [PMID: 32933106 PMCID: PMC7565890 DOI: 10.3390/cells9092090] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022] Open
Abstract
The airway epithelium contains ionocytes, a rare cell type with high expression of Forkhead Box I1 (FOXI1) transcription factor and Cystic Fibrosis Transmembrane conductance Regulator (CFTR), a chloride channel that is defective in cystic fibrosis (CF). Our aim was to verify if ionocyte development is altered in CF and to investigate the relationship between ionocytes and CFTR-dependent chloride secretion. We collected nasal cells by brushing to determine ionocyte abundance. Nasal and bronchial cells were also expanded in vitro and reprogrammed to differentiated epithelia for morphological and functional studies. We found a relatively high (~3%) ionocyte abundance in ex vivo nasal samples, with no difference between CF and control individuals. In bronchi, ionocytes instead appeared very rarely as previously reported, thus suggesting a possible proximal-distal gradient in human airways. The difference between nasal and bronchial epithelial cells was maintained in culture, which suggests an epigenetic control of ionocyte development. In the differentiation phase of the culture procedure, we used two media that resulted in a different pattern of CFTR expression: confined to ionocytes or more broadly expressed. CFTR function was similar in both conditions, thus indicating that chloride secretion equally occurs irrespective of CFTR expression pattern.
Collapse
Affiliation(s)
- Paolo Scudieri
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genova, 16147 Genova, Italy; (P.S.); (I.M.)
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Ilaria Musante
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genova, 16147 Genova, Italy; (P.S.); (I.M.)
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Arianna Venturini
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Michele Genovese
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
| | - Federico Cresta
- Centro Fibrosi Cistica, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Emanuela Caci
- Medical Genetics Unit, Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Alessandro Palleschi
- Thoracic Surgery and Lung Transplantation Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Marco Poeta
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
| | - Francesca Santamaria
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
| | - Fabiana Ciciriello
- Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, 00165 Roma, Italy; (F.C.); (V.L.)
| | - Vincenzina Lucidi
- Cystic Fibrosis Unit, Bambino Gesù Children’s Hospital, 00165 Roma, Italy; (F.C.); (V.L.)
| | - Luis J. V. Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli (NA), Italy; (A.V.); (D.G.); (M.G.)
- Department of Translational Medical Sciences, Università di Napoli “Federico II”, 80131 Napoli, Italy; (M.P.); (F.S.)
- Correspondence:
| |
Collapse
|
34
|
Wirsching E, Fauler M, Fois G, Frick M. P2 Purinergic Signaling in the Distal Lung in Health and Disease. Int J Mol Sci 2020; 21:E4973. [PMID: 32674494 PMCID: PMC7404078 DOI: 10.3390/ijms21144973] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
The distal lung provides an intricate structure for gas exchange in mammalian lungs. Efficient gas exchange depends on the functional integrity of lung alveoli. The cells in the alveolar tissue serve various functions to maintain alveolar structure, integrity and homeostasis. Alveolar epithelial cells secrete pulmonary surfactant, regulate the alveolar surface liquid (ASL) volume and, together with resident and infiltrating immune cells, provide a powerful host-defense system against a multitude of particles, microbes and toxicants. It is well established that all of these cells express purinergic P2 receptors and that purinergic signaling plays important roles in maintaining alveolar homeostasis. Therefore, it is not surprising that purinergic signaling also contributes to development and progression of severe pathological conditions like pulmonary inflammation, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pulmonary fibrosis. Within this review we focus on the role of P2 purinergic signaling in the distal lung in health and disease. We recapitulate the expression of P2 receptors within the cells in the alveoli, the possible sources of ATP (adenosine triphosphate) within alveoli and the contribution of purinergic signaling to regulation of surfactant secretion, ASL volume and composition, as well as immune homeostasis. Finally, we summarize current knowledge of the role for P2 signaling in infectious pneumonia, ALI/ARDS and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
| | | | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (E.W.); (M.F.); (G.F.)
| |
Collapse
|
35
|
Molinari G, Molinari L, Nervo E. Environmental and Endogenous Acids Can Trigger Allergic-Type Airway Reactions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4688. [PMID: 32610702 PMCID: PMC7370125 DOI: 10.3390/ijerph17134688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/20/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Inflammatory allergic and nonallergic respiratory disorders are spreading worldwide and often coexist. The root cause is not clear. This review demonstrates that, from a biochemical point of view, it is ascribable to protons (H+) released into cells by exogenous and endogenous acids. The hypothesis of acids as the common cause stems from two considerations: (a) it has long been known that exogenous acids present in air pollutants can induce the irritation of epithelial surfaces, particularly the airways, inflammation, and bronchospasm; (b) according to recent articles, endogenous acids, generated in cells by phospholipases, play a key role in the biochemical mechanisms of initiation and progression of allergic-type reactions. Therefore, the intracellular acidification and consequent Ca2+ increase, induced by protons generated by either acid pollutants or endogenous phospholipases, may constitute the basic mechanism of the multimorbidity of these disorders, and environmental acidity may contribute to their spread.
Collapse
Affiliation(s)
- Giuliano Molinari
- Studio Tecnico Ing. Laura Molinari, Environmental Health and Safety Via Quarto Ponte 17, 37138 Verona, Italy;
| | - Laura Molinari
- Studio Tecnico Ing. Laura Molinari, Environmental Health and Safety Via Quarto Ponte 17, 37138 Verona, Italy;
| | - Elsa Nervo
- Elsa Nervo, Società Chimica Italiana, 00198 Rome, Italy;
| |
Collapse
|
36
|
Zhang H, Cui Y, Zhou Z, Ding Y, Nie H. Alveolar Type 2 Epithelial Cells as Potential Therapeutics for Acute Lung Injury/Acute Respiratory Distress Syndrome. Curr Pharm Des 2020; 25:4877-4882. [PMID: 31801451 DOI: 10.2174/1381612825666191204092456] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Acute lung injury/acute respiratory distress syndrome is a common clinical illness with high morbidity and mortality, which is still one of the medical problems urgently needed to be solved. Alveolar type 2 epithelial cells are an important component of lung epithelial cells and as a kind of stem cells, they can proliferate and differentiate into alveolar type 1 epithelial cells, thus contributing to lung epithelial repairment. In addition, they synthesize and secrete all components of the surfactant that regulates alveolar surface tension in the lungs. Moreover, alveolar type 2 epithelial cells play an active role in enhancing alveolar fluid clearance and reducing lung inflammation. In recent years, as more advanced approaches appear in the field of stem and progenitor cells in the lung, many preclinical studies have shown that the cell therapy of alveolar type 2 epithelial cells has great potential effects for acute lung injury/acute respiratory distress syndrome. We reviewed the recent progress on the mechanisms of alveolar type 2 epithelial cells involved in the damaged lung repairment, aiming to explore the possible therapeutic targets in acute lung injury/acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
37
|
Measurement of Multi Ion Transport through Human Bronchial Epithelial Cell Line Provides an Insight into the Mechanism of Defective Water Transport in Cystic Fibrosis. MEMBRANES 2020; 10:membranes10030043. [PMID: 32178452 PMCID: PMC7142439 DOI: 10.3390/membranes10030043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 12/28/2022]
Abstract
We measured concentration changes of sodium, potassium, chloride ions, pH and the transepithelial potential difference by means of ion-selective electrodes, which were placed on both sides of a human bronchial epithelial 16HBE14σ cell line grown on a porous support in the presence of ion channel blockers. We found that, in the isosmotic transepithelial concentration gradient of either sodium or chloride ions, there is an electroneutral transport of the isosmotic solution of sodium chloride in both directions across the cell monolayer. The transepithelial potential difference is below 3 mV. Potassium and pH change plays a minor role in ion transport. Based on our measurements, we hypothesize that in a healthy bronchial epithelium, there is a dynamic balance between water absorption and secretion. Water absorption is caused by the action of two exchangers, Na/H and Cl/HCO3, secreting weakly dissociated carbonic acid in exchange for well dissociated NaCl and water. The water secretion phase is triggered by an apical low volume-dependent factor opening the Cystic Fibrosis Transmembrane Regulator CFTR channel and secreting anions that are accompanied by paracellular sodium and water transport.
Collapse
|
38
|
Small Molecule Anion Carriers Correct Abnormal Airway Surface Liquid Properties in Cystic Fibrosis Airway Epithelia. Int J Mol Sci 2020; 21:ijms21041488. [PMID: 32098269 PMCID: PMC7073096 DOI: 10.3390/ijms21041488] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 01/03/2023] Open
Abstract
Cystic fibrosis (CF) is a genetic disease characterized by the lack of cystic fibrosis transmembrane conductance regulator (CFTR) protein expressed in epithelial cells. The resulting defective chloride and bicarbonate secretion and imbalance of the transepithelial homeostasis lead to abnormal airway surface liquid (ASL) composition and properties. The reduced ASL volume impairs ciliary beating with the consequent accumulation of sticky mucus. This situation prevents the normal mucociliary clearance, favouring the survival and proliferation of bacteria and contributing to the genesis of CF lung disease. Here, we have explored the potential of small molecules capable of facilitating the transmembrane transport of chloride and bicarbonate in order to replace the defective transport activity elicited by CFTR in CF airway epithelia. Primary human bronchial epithelial cells obtained from CF and non-CF patients were differentiated into a mucociliated epithelia in order to assess the effects of our compounds on some key properties of ASL. The treatment of these functional models with non-toxic doses of the synthetic anionophores improved the periciliary fluid composition, reducing the fluid re-absorption, correcting the ASL pH and reducing the viscosity of the mucus, thus representing promising drug candidates for CF therapy.
Collapse
|
39
|
Abstract
The respiratory system plays an essential role for human life. This system (like all others) undergoes physiological regeneration due to many types of stem cells found both in the respiratory tract itself and in the alveoli. The stem cell hierarchy is very extensive due to their variety in the lungs and is still not completely understood.The best described lung stem cells are alveolar type II cells, which as progenitor lung stem cells are precursors of alveolar type I cells, i.e., cells that perform gas exchange in the lungs. These progenitor stem cells, which reside in alveoli corners, express high levels of surfactant protein C (SFTPC). Despite the fact that type II pneumocytes occupy only 7-10% of the lung surface, there are almost twice as many as alveolar type I cells occupying almost 95% of the surface.Other stem cells making up the lung regenerative potential have also been identified in the lungs. Both endothelial, mesodermal, and epithelial stem cells are necessary for the lungs to function properly and perform their physiological functions.The lungs, like all other organs, undergo an aging process. As a result of this process, not only the total number of cells changes, the percentage of particular types of cells, but also their efficiency is reduced. With age, the proliferative potential of lung stem cells also decreases, not just their number. This brings about the need to increase the intensity of research in the field of regenerative medicine.
Collapse
Affiliation(s)
- Andrzej Ciechanowicz
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical Univeristy of Warsaw, Warsaw, Poland.
| |
Collapse
|
40
|
Zhang Y, MacKenzie B, Koleng JJ, Maier E, Warnken ZN, Williams RO. Development of an Excipient-Free Peptide Dry Powder Inhalation for the Treatment of Pulmonary Fibrosis. Mol Pharm 2020; 17:632-644. [PMID: 31913640 DOI: 10.1021/acs.molpharmaceut.9b01085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The caveolin scaffolding domain peptide (CSP) is being developed for the therapeutic intervention of a lethal lung disease, idiopathic pulmonary fibrosis. While direct respiratory delivery of CSP7 (a 7-mer fragment of CSP) is considered an effective route, proper formulation and processing of the peptide are required. First, air-jet milling technology was performed in order to micronize the neat peptide powder. Next, the fine particles were subjected to a stability study with physical and chemical characterizations. In addition, the in vivo efficacy of processed CSP7 powder was evaluated in an animal model of lung fibrosis. The results revealed that, with jet milling, the particle size of CSP7 was reduced to a mass median aerodynamic diameter of 1.58 ± 0.1 μm and 93.3 ± 3.3% fine particle fraction, optimal for deep lung delivery. A statistically significant reduction of collagen was observed in diseased lung tissues of mice that received CSP7 powder for inhalation. The particles remained chemically and physically stable after micronization and during storage. This work demonstrated that jet milling is effective in the manufacturing of a stable, excipient-free CSP7 inhalation powder for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Yajie Zhang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , 2409 University Avenue , Austin , Texas 78712 , United States
| | - BreAnne MacKenzie
- Lung Therapeutics Inc. , 2600 Via Fortuna, Suite 360 , Austin , Texas 78746 , United States
| | - John J Koleng
- Lung Therapeutics Inc. , 2600 Via Fortuna, Suite 360 , Austin , Texas 78746 , United States
| | - Esther Maier
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , 2409 University Avenue , Austin , Texas 78712 , United States
| | - Zachary N Warnken
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , 2409 University Avenue , Austin , Texas 78712 , United States
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy , The University of Texas at Austin , 2409 University Avenue , Austin , Texas 78712 , United States
| |
Collapse
|
41
|
Wang J, Luo J, Huang W, Liu C, Zeng D, Liu H, Qu X, Liu C, Xiang Y, Qin X. Increased intracellular Cl - concentration by activating FAK promotes airway epithelial BEAS-2B cells proliferation and wound healing. Arch Biochem Biophys 2019; 680:108225. [PMID: 31838119 DOI: 10.1016/j.abb.2019.108225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/10/2019] [Accepted: 12/10/2019] [Indexed: 11/30/2022]
Abstract
An increase in intracellular Cl- concentration ([Cl-]i) may be a general response of airway epithelial cells to various stimuli and may participate in some basic cellular functions. However, whether the basic functional activities of cells, such as proliferation and wound healing, are related to Cl- activities remains unclear. This study aimed to investigate the effects and potential mechanisms of [Cl-]i on the proliferation and wound healing ability of airway epithelial BEAS-2B cells. BEAS-2B cells were treated with four Cl- channel inhibitors (T16Ainh-A01, CFTRinh-172, CaCCinh-A01, and IAA-94), and the Cl- fluorescence probe N-(ethoxycarbonylmethyl)-6-methoxyquinolinium bromide was used. Results showed that all Cl- channel inhibitors could increase [Cl-]i in BEAS-2B cells. The increased [Cl-]i induced by Cl- channel inhibitors or clamping [Cl-]i at high levels enhanced the phosphorylation of focal adhesion kinase (FAK) and subsequently promoted the proliferation and wound healing ability of BEAS-2B cells. By contrast, the FAK inhibitor PF573228 abrogated these effects induced by the increased [Cl-]i. FAK also activated the PI3K/AKT signaling pathway. In conclusion, increased [Cl-]i promotes the proliferation and wound healing ability of BEAS-2B cells by activating FAK to activate the PI3K/AKT signaling pathway. Intracellular Cl- may act as a signaling molecule to regulate the proliferation and wound healing ability of airway epithelial cells.
Collapse
Affiliation(s)
- Jia Wang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China; Hunan Provincial People's Hospital, The First-affiliated Hospital of Hunan Normal University, Changsha, 410016, China
| | - Jinhua Luo
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China
| | - Wenjie Huang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China
| | - Caixia Liu
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Dan Zeng
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China; Hunan Provincial People's Hospital, The First-affiliated Hospital of Hunan Normal University, Changsha, 410016, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China.
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine, Central South University, Changsha, 410000, China.
| |
Collapse
|
42
|
Zellnitz S, Zellnitz L, Müller M, Meindl C, Schröttner H, Fröhlich E. Impact of drug particle shape on permeability and cellular uptake in the lung. Eur J Pharm Sci 2019; 139:105065. [DOI: 10.1016/j.ejps.2019.105065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/24/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023]
|
43
|
Öztop M, Özbek M, Liman N, Beyaz F, Ergün E, Ergün L, Kavraal UK, Ergen E. Expression patterns of natriuretic peptides in pre-hibernating and hibernating anatolian ground squirrel (Spermophilus xanthoprymnus) lung. Acta Histochem 2019; 121:852-865. [PMID: 31445760 DOI: 10.1016/j.acthis.2019.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/16/2019] [Accepted: 08/18/2019] [Indexed: 12/14/2022]
Abstract
Anatolian ground squirrel (Spermophilus xanthoprymnus) is a true hibernator. This animal transiently reduces pulmonary function during hibernation. Continuance of pulmonary function is very important to survive ground squirrels during the hibernation. Natriuretic peptides may be key players in the modulation of pulmonary hemostasis. However, NPs' role in pulmonary function during hibernation remains unclear. We aimed to investigate the localization and distribution of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and C-type natriuretic peptide (CNP) in squirrel lungs during pre-hibernation and hibernation periods using immunohistochemistry. Our immunohistochemical data indicate that ANP, BNP, and CNP were produced by the mucosal epithelium of terminal and respiratory bronchioles, smooth muscle cells in the lamina propria of terminal bronchioles and vascular smooth muscle cells, alveolar type II cells, and macrophages. ANP immunoreactivity was weaker than BNP and CNP immunoreactivities in these cells. The results also demonstrate that the number of ANP, BNP and CNP positive alveolar type II cells tended to increase, although statistically non-significant, during the hibernation period, but the expression of NPs in other pulmonary cells is unaffected by hibernation. This study firstly investigates ANP, BNP and CNP distribution in the Anatolian ground squirrel lung. However, further studies are required to dissect their functional roles during the hibernation.
Collapse
|
44
|
Huang S, Constant S, De Servi B, Meloni M, Culig J, Bertini M, Saaid A. In vitro safety and performance evaluation of a seawater solution enriched with copper, hyaluronic acid, and eucalyptus for nasal lavage. MEDICAL DEVICES-EVIDENCE AND RESEARCH 2019; 12:399-410. [PMID: 31576180 PMCID: PMC6766585 DOI: 10.2147/mder.s209644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/20/2019] [Indexed: 01/02/2023] Open
Abstract
Background The common cold is a viral infectious disease with symptoms such as runny nose, sore throat, and mainly, nasal congestion. State-of-the-art therapeutic approaches focus on alleviating the symptoms of this disease by non-invasive and simple-to-use methods. Nasal irrigation is one of the most accepted approaches to ease nasal congestion which, if left untreated, has a negative impact on the quality of life of patients. Purpose In this study, the safety and efficacy of a novel hypertonic seawater solution for nasal lavage enriched with hyaluronic acids, eucalyptus oil, copper, and manganese salts (Stérimar Stop & Protect Cold and Flu; SSPCF) have been investigated in vitro. Methods An in vitro 3D reconstituted human nasal epithelium tissue model, MucilAir™, has been used in this study to investigate the safety of SSPCF on nasal epithelium by measuring transepithelial electrical resistance (TEER), lactate dehydrogenase (LDH), and interleukin-8 (IL-8) secretion. The efficacy of SSPCF was measured by mucociliary clearance (MCC), ATP release, Alcian blue and aquaporin (AQP3) stainings. Results SSPCF treatment respected nasal epithelium tissue integrity and enhanced barrier function without inducing a cytotoxic response. Secreted LDH and IL-8 levels were similar to untreated controls. MCC rate was increased 2.5-fold and ATP release decreased 87% upon SSPCF treatment, indicating improved decongestion activity. SSPCF treatment after hypotonic stress helped recover cellular organization, as shown by Alcian blue and AQP3 staining assays. Conclusion SSPCF appears as a safe and effective nasal irrigation formula that may alleviate the symptoms associated with common cold such as nasal congestion.
Collapse
Affiliation(s)
| | | | - Barbara De Servi
- Department of in Vitro Research, VitroScreen, Milan, 20149, Italy
| | - Marisa Meloni
- Department of in Vitro Research, VitroScreen, Milan, 20149, Italy
| | - Josip Culig
- Department of Pharmacology, University of Applied Health Sciences, Zagreb, 10000, Croatia
| | - Marco Bertini
- R&D Department, Laboratori Baldacci SpA, Pisa, Italy
| | - Amina Saaid
- Department of R&D and Innovation, Laboratoire Fumouze, Levallois-Perret, 92686, France
| |
Collapse
|
45
|
A physiologically-motivated model of cystic fibrosis liquid and solute transport dynamics across primary human nasal epithelia. J Pharmacokinet Pharmacodyn 2019; 46:457-472. [PMID: 31494805 DOI: 10.1007/s10928-019-09649-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/02/2019] [Indexed: 11/27/2022]
Abstract
Cystic fibrosis (CF) disease is caused by mutations affecting the gene coding for the cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel expressed in the mucosal side of epithelial tissue. In the airway, dysfunctional CFTR results in a transepithelial osmotic imbalance leading to hyperabsorption of airway surface liquid mucostasis, chronic inflammation, and eventual respiratory failure. Human nasal epithelial cell cultures from healthy and CF donors were used to perform studies of liquid and solute transport dynamics at an air/liquid interface in order to emulate the in vivo airway. Then, these results were used to inform a quantitative systems pharmacology model of airway epithelium describing electrically and chemically driven transcellular ionic transport, contributions of both convective and diffusive paracellular solute transport, and osmotically driven transepithelial water dynamics. Model predictions showed CF cultures, relative to non-CF ones, have increased apical and basolateral water permeabilities, and increase paracellular permeability and transepithelial chemical driving force for a radiolabeled tracer used to track small molecule absorption. These results provide a computational platform to better understand and probe the mechanisms behind the liquid hyperabsorption and small molecule retention profiles observed in the CF airway.
Collapse
|
46
|
Turck D, Castenmiller J, de Henauw S, Hirsch-Ernst KI, Kearney J, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Aggett P, Fairweather-Tait S, Martin A, Przyrembel H, de Sesmaisons-Lecarré A, Naska A. Dietary reference values for chloride. EFSA J 2019; 17:e05779. [PMID: 32626426 PMCID: PMC7009052 DOI: 10.2903/j.efsa.2019.5779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) has derived dietary reference values (DRVs) for chloride. There are no appropriate biomarkers of chloride status, no balance studies and no adequate evidence on the relationship between chloride intake and health outcomes that can be used to set DRVs for chloride. There is a close relationship between sodium and chloride balances in the body. Sodium chloride is the main source of both electrolytes in European diets and similar urinary excretion levels of sodium and chloride (on a molar basis) are typically observed in Western populations. Hence, the Panel considered that reference values for chloride can be set at values equimolar to the reference values for sodium for all population groups, and are as follows: 1.7 g/day for children aged 1-3 years, 2.0 g/day for children aged 4-6 years, 2.6 g/day for children aged 7-10 years, 3.1 g/day for children aged 11-17 years and 3.1 g/day for adults including pregnant and lactating women. Consistent with the reference values for sodium, these levels of chloride intake are considered to be safe and adequate for the general EU population, under the consideration that the main dietary source of chloride intake is sodium chloride. For infants aged 7-11 months, an adequate intake of 0.3 g/day is set.
Collapse
|
47
|
Scheiber IF, Alarcon NO, Zhao N. Manganese Uptake by A549 Cells is Mediated by Both ZIP8 and ZIP14. Nutrients 2019; 11:nu11071473. [PMID: 31261654 PMCID: PMC6682971 DOI: 10.3390/nu11071473] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022] Open
Abstract
The alveolar epithelia of the lungs require manganese (Mn) as an essential nutrient, but also provide an entry route for airborne Mn that can cause neurotoxicity. Transporters involved in Mn uptake by alveolar epithelial cells are unknown. Recently, two members of the Zrt- and Irt-like protein (ZIP) family of metal transporters, ZIP8 and ZIP14, have been identified as crucial Mn importers in vivo. ZIP8 is by far most abundantly expressed in the lungs, whereas ZIP14 expression in the lungs is low compared to other tissues. We hypothesized that Mn uptake by alveolar epithelial cells is primarily mediated by ZIP8. To test our hypothesis, we used A549 cells, a type II alveolar cell line. Mirroring the in vivo situation, A549 cells expressed higher levels of ZIP8 than cell models for the liver, intestines, and kidney. Quantification of ZIP8 and ZIP14 revealed a strong enrichment of ZIP8 over ZIP14 in A549 cells. Using siRNA technology, we identified ZIP8 and ZIP14 as the major transporters mediating Mn uptake by A549 cells. To our surprise, knockdown of either ZIP8 or ZIP14 impaired Mn accumulation to a similar extent, which we traced back to similar amounts of ZIP8 and ZIP14 at the plasma membrane. Our study highlights the importance of both ZIP8 and ZIP14 in Mn metabolism of alveolar epithelial cells.
Collapse
Affiliation(s)
- Ivo F Scheiber
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA
| | | | - Ningning Zhao
- Department of Nutritional Sciences, The University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
48
|
New ISE-Based Apparatus for Na +, K +, Cl -, pH and Transepithelial Potential Difference Real-Time Simultaneous Measurements of Ion Transport across Epithelial Cells Monolayer⁻Advantages and Pitfalls. SENSORS 2019; 19:s19081881. [PMID: 31009998 PMCID: PMC6515391 DOI: 10.3390/s19081881] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/14/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022]
Abstract
Cystic Fibrosis (CF) is the most common fatal human genetic disease, which is caused by a defect in an anion channel protein (CFTR) that affects ion and water transport across the epithelium. We devised an apparatus to enable the measurement of concentration changes of sodium, potassium, chloride, pH, and transepithelial potential difference by means of ion-selective electrodes that were placed on both sides of a 16HBE14σ human bronchial epithelial cell line that was grown on a porous support. Using flat miniaturized ISE electrodes allows for reducing the medium volume adjacent to cells to approximately 20 μL and detecting changes in ion concentrations that are caused by transport through the cell layer. In contrast to classic electrochemical measurements, in our experiments neither the calibration of electrodes nor the interpretation of results is simple. The calibration solutions might affect cell physiology, the medium composition might change the direction of actions of the membrane channels and transporters, and water flow that might trigger or cut off the transport pathways accompanies the transport of ions. We found that there is an electroneutral transport of sodium chloride in both directions of the cell monolayer in the isosmotic transepithelial concentration gradient of sodium or chloride ions. The ions and water are transported as an isosmotic solution of 145 mM of NaCl.
Collapse
|
49
|
Lu K, Chen X, Zhu W, Mao X, Yang Y, Qiu J, Zhang M, Cheng R. Terbutaline alleviates the lung injury in the neonatal rats exposed to endotoxin: Potential roles of epithelial sodium channels. Pediatr Pulmonol 2019; 54:280-288. [PMID: 30644180 PMCID: PMC6618278 DOI: 10.1002/ppul.24242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/15/2018] [Indexed: 12/18/2022]
Abstract
Intrauterine inflammation generates inflammatory mediators that damage the developing bronchoalveolar epithelium, resulting in neonatal lung injury. Lung fluid transport disorders are the main reasons for the development of pulmonary edema, an important pathology of lung injury. Previous studies suggested that epithelial sodium channels (ENaCs) play an important role in lung fluid transport. Here, we investigated whether changes in the expression of ENaCs were observed when neonatal rat lung injury was induced by maternal exposure to endotoxin. We also examined the therapeutic effect of terbutaline nebulizer inhalation on this injury. The results showed that maternal exposure to endotoxin increased the levels of TNF-α and IL-1β in bronchoalveolar lavage fluid, suppressed α-, β-, γ-ENaC in the neonatal rat lung, and resulted in the formation of pulmonary edema on postnatal days 1 and 7. Terbutaline up-regulated the expression of β- and γ-ENaC in the distal lung after 7 days of treatment. The potential signal molecules cAMP, PKA, and CREB expressions were increased after terbutaline treatment. In summary, maternal exposure to endotoxin decreased the expression of ENaCs in neonatal rats which, in turn, may exacerbate pulmonary edema. Inhalation of the β2-adrenergic receptor agonist terbutaline improved lung liquid clearance. By increasing the expression of sodium ion channels, the effective removal of alveolar fluid provides a new way for the prevention and treatment of neonatal lung injury.
Collapse
Affiliation(s)
- Keyu Lu
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Zhu
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaonan Mao
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Qiu
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, China.,Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
50
|
Abstract
Mucociliary clearance is critically important in protecting the airways from infection and from the harmful effects of smoke and various inspired substances known to induce oxidative stress and persistent inflammation. An essential feature of the clearance mechanism involves regulation of the periciliary liquid layer on the surface of the airway epithelium, which is necessary for normal ciliary beating and maintenance of mucus hydration. The underlying ion transport processes associated with airway surface hydration include epithelial Na+ channel-dependent Na+ absorption occurring in parallel with CFTR and Ca2+-activated Cl- channel-dependent anion secretion, which are coordinately regulated to control the depth of the periciliary liquid layer. Oxidative stress is known to cause both acute and chronic effects on airway ion transport function, and an increasing number of studies in the past few years have identified an important role for autophagy as part of the physiological response to the damaging effects of oxidation. In this review, recent studies addressing the influence of oxidative stress and autophagy on airway ion transport pathways, along with results showing the potential of autophagy modulators in restoring the function of ion channels involved in transepithelial electrolyte transport necessary for effective mucociliary clearance, are presented.
Collapse
Affiliation(s)
- Scott M O'Grady
- Departments of Animal Science, Integrative Biology and Physiology, University of Minnesota , St. Paul, Minnesota
| |
Collapse
|