1
|
Zheng YB, Lu S, Chu TB, Pang GF, Yang LY, Zhang Q. Investigate the potential impact of Hemagglutinin from the H1N1 strain on severe pneumonia. Gene 2024; 926:148559. [PMID: 38740352 DOI: 10.1016/j.gene.2024.148559] [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: 01/20/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
The most prevalent glycoprotein on the influenza virus envelope is called hemagglutinin (HA), yet little is known about its involvement in the pathophysiology and etiology of severe influenza pneumonia. Here, after stimulating human bronchial epithelial cells (16-HBE) and mice with HA of H1N1 for 12 h, we investigated the proliferation, migration, inflammatory cytokines expression, and apoptosis in 16-HBE and the pathological damage in mouse lung tissue. The expression of inflammatory cytokines plasminogen activator inhibitor 1(PAI-1), urokinase-type (uPA) and tissue-type (tPA) plasminogen activators, and apoptosis were all enhanced by HA, which also prevented the proliferation and migration of bronchial epithelial cells. HA enhanced up-regulated PAI-1, uPA, and tPA protein expression within mouse lung tissue and caused lung injury. In conclusion, HA alone, but not the whole H1N1 virus, induces lung tissue injury by inhibiting cell proliferation and migration, while promoting the expression of inflammatory cytokines and apoptosis.
Collapse
Affiliation(s)
- Yu-Bi Zheng
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China.
| | - Song Lu
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Tian-Bao Chu
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Gui-Feng Pang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Lin-Ying Yang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China
| | - Qing Zhang
- Affiliated Hospital of Chengde Medical University, Department of Respiratory and Critical Care Medicine, Chengde 067000, Hebei, China.
| |
Collapse
|
2
|
Lee N, Hwang YJ, Na HG, Cho DH. Far-infrared irradiation inhibits proliferation of human upper airway epithelial cells via protein phosphatase 2A-promoted dephosphorylation of p70 S6 kinase. Photochem Photobiol Sci 2024:10.1007/s43630-024-00652-0. [PMID: 39461912 DOI: 10.1007/s43630-024-00652-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Far-infrared (FIR) ray, an invisible electromagnetic radiation with a wavelength of 3‒1000 μm, elicits various biological effects. Excessive proliferation of human upper airway epithelial cells (HUAEpCs) contributes to the development and exacerbation of nasal narrowing diseases, including nasal polyposis and chronic rhinosinusitis with nasal polyps (CRSwNP). Here, we investigated the molecular mechanisms through which FIR irradiation inhibits the proliferation of HUAEpCs. FIR irradiation significantly inhibited the proliferation of NCI-H292 cells without alteration in cell viability. The anti-proliferative effect of FIR radiation was accompanied by decreased phosphorylation of p70S6K at Thr389 (p-p70S6K-Thr389), without changes in the phosphorylation of mammalian target of rapamycin and adenosine monophosphate-activated protein kinase (AMPK). Overexpression of p70S6K-T389E mutant gene, not dominant negative-AMPKα1 gene, significantly reversed FIR irradiation-inhibited p-p70S6K-Thr389 and cell proliferation. Cotreatment with okadaic acid or knockdown of protein phosphatase 2A catalytic subunit (PP2Ac) gene expression significantly reversed FIR irradiation-decreased p-p70S6K-Thr389 and cell proliferation. FIR irradiation remarkably promoted the physical association of p70S6K and PP2Ac without change in total PP2Ac expression. Hyperthermal stimulus (39 °C) did not alter p-p70S6K-Thr389 and cell proliferation. In line with NCI-H292 cell results, FIR irradiation, not hyperthermal stimulus, significantly decreased p-p70S6K-Thr389 and cell proliferation in primary human nasal turbinate and polyp epithelial cells. These results demonstrated that FIR irradiation decreased the proliferation of HUAEpCs through PP2A-mediated inhibition of p70S6K phosphorylation, independent of its hyperthermal effect. Our data suggest that FIR therapy can be used to treat upper airway narrowing epithelial hyperplastic diseases, including nasal polyposis and CRSwNP.
Collapse
Affiliation(s)
- Nayoung Lee
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea
| | - Yun-Jin Hwang
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea
| | - Hyung Gyun Na
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea
| | - Du-Hyong Cho
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, South Korea.
| |
Collapse
|
3
|
Qiao Y, Guo Y, Wang X, Zhang W, Guo W, Wang Z, Liu C. Multi-omics analysis reveals the enhancing effects of Glycyrrhiza polysaccharides on the respiratory health of broilers. Int J Biol Macromol 2024; 280:135953. [PMID: 39322162 DOI: 10.1016/j.ijbiomac.2024.135953] [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: 12/22/2023] [Revised: 09/14/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
This study investigated the impact of Glycyrrhiza polysaccharides (GPS) on the respiratory health of broilers. Specifically, 240 one-day-old male Arbor Acres (AA) broilers were randomly assigned to two groups: basal diet (CON) and GPS (supplemented with 150 mg/kg of Glycyrrhiza polysaccharides). When compared with the CON group, the GPS group significantly increased the broiler average daily gain, serum immunoglobulin A, immunoglobulin M, immunoglobulin G, antioxidant capacity, superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and tracheal messenger RNA (mRNA) expression levels of SOD1, SOD2, and GSH-Px. The GPS group also had a reduced feed conversion ratio, reduced lung IL-1β and IL-6 levels, and upregulated tracheal mRNA expression of Occludin, Claudin1, and Mucin-2. Additionally, the GPS group had alterations in lung microbial diversity and composition. Transcriptomic and metabolomic analyses revealed the activation of the T cell receptor (TCR) signaling pathway and linoleic acid metabolic pathway in the GPS group. Correlation analysis demonstrated significant associations between differential bacteria, genes, serum metabolites, and phenotypic indicators. In conclusion, Glycyrrhiza polysaccharide supplementation positively influenced the respiratory health of broilers by modulating the lung microbiota, activating the TCR signaling pathway, and affecting the linoleic acid metabolism pathway.
Collapse
Affiliation(s)
- Yingying Qiao
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China; College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yongpeng Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Xinlei Wang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China
| | - Wei Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Weibing Guo
- Inner Mongolia Evergrand Pharmaceutical Co. LTD, Chifeng 025250, China
| | - Zhixiang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China.
| | - Changzhong Liu
- College of Animal Science, Henan Institute of Science and Technology, Xinxiang 453003, China.
| |
Collapse
|
4
|
de Hilster RHJ, Reinders-Luinge MA, Schuil A, Borghuis T, Harmsen MC, Burgess JK, Hylkema MN. A 3D Epithelial-Mesenchymal Co-Culture Model of the Airway Wall Using Native Lung Extracellular Matrix. Bioengineering (Basel) 2024; 11:946. [PMID: 39329688 PMCID: PMC11428669 DOI: 10.3390/bioengineering11090946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterized by ongoing inflammation, impaired tissue repair, and aberrant interplay between airway epithelium and fibroblasts, resulting in an altered extracellular matrix (ECM) composition. The ECM is the three-dimensional (3D) scaffold that provides mechanical support and biochemical signals to cells, now recognized not only as a consequence but as a potential driver of disease progression. To elucidate how the ECM influences pathophysiological changes occurring in COPD, in vitro models are needed that incorporate the ECM. ECM hydrogels are a novel experimental tool for incorporating the ECM in experimental setups. We developed an airway wall model by combining lung-derived ECM hydrogels with a co-culture of primary human fibroblasts and epithelial cells at an air-liquid interface. Collagen IV and a mixture of collagen I, fibronectin, and bovine serum albumin were used as basement membrane-mimicking coatings. The model was initially assembled using porcine lung-derived ECM hydrogels and subsequently with COPD and non-COPD human lung-derived ECM hydrogels. The resulting 3D construct exhibited considerable contraction and supported co-culture, resulting in a differentiated epithelial layer. This multi-component 3D model allows the investigation of remodelling mechanisms, exploring ECM involvement in cellular crosstalk, and holds promise as a model for drug discovery studies exploring ECM involvement in cellular interactions.
Collapse
Affiliation(s)
- Roderick H. J. de Hilster
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Marjan A. Reinders-Luinge
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Annemarie Schuil
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Theo Borghuis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Martin C. Harmsen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Machteld N. Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
5
|
Imayama I, Eccles JD, Ascoli C, Kudlaty E, Park GY. Body Weight and Allergic Asthma: A Narrative Review. J Clin Med 2024; 13:4801. [PMID: 39200943 PMCID: PMC11355285 DOI: 10.3390/jcm13164801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Obesity is a known risk factor for asthma development, progression, and exacerbation. Nevertheless, the underlying pathophysiological mechanisms explaining how obesity contributes to the development and progression of asthma have yet to be established. Here, we review human studies examining the associations between asthma and obesity, focusing on the literature from the past 10 years. Overall, current evidence suggests that while both asthma and obesity are complex diseases with significant heterogeneity, they both share various features of chronic inflammation. Furthermore, the interactions between asthma and obesity likely involve allergen-specific T helper type 2 (type 2) immune responses, as well as diverse non-type 2 inflammatory pathways. However, despite considerable progress, studies to date have not definitively elucidated the mechanisms that account for the observed association. A large-scale population-based study combined with translational immunological research, including targeted asthma therapies and pharmacological weight loss therapies, may be required to properly dissect the details of obesity-related asthma pathophysiology.
Collapse
Affiliation(s)
- Ikuyo Imayama
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL 60612, USA; (J.D.E.); (C.A.); (E.K.); (G.Y.P.)
- StatCare, Knoxville, TN 37919, USA
| | - Jacob D. Eccles
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL 60612, USA; (J.D.E.); (C.A.); (E.K.); (G.Y.P.)
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL 60612, USA; (J.D.E.); (C.A.); (E.K.); (G.Y.P.)
| | - Elizabeth Kudlaty
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL 60612, USA; (J.D.E.); (C.A.); (E.K.); (G.Y.P.)
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL 60612, USA; (J.D.E.); (C.A.); (E.K.); (G.Y.P.)
| |
Collapse
|
6
|
Sharma S, Gerber AN, Kraft M, Wenzel SE. Asthma Pathogenesis: Phenotypes, Therapies, and Gaps: Summary of the Aspen Lung Conference 2023. Am J Respir Cell Mol Biol 2024; 71:154-168. [PMID: 38635858 PMCID: PMC11299090 DOI: 10.1165/rcmb.2024-0082ws] [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] [Received: 02/21/2024] [Accepted: 04/17/2024] [Indexed: 04/20/2024] Open
Abstract
Although substantial progress has been made in our understanding of asthma pathogenesis and phenotypes over the nearly 60-year history of the Aspen Lung Conferences on asthma, many ongoing challenges exist in our understanding of the clinical and molecular heterogeneity of the disease and an individual patient's response to therapy. This report summarizes the proceedings of the 2023 Aspen Lung Conference, which was organized to review the clinical and molecular heterogeneity of asthma and to better understand the impact of genetic, environmental, cellular, and molecular influences on disease susceptibility, heterogeneity, and severity. The goals of the conference were to review new information about asthma phenotypes, cellular processes, and cellular signatures underlying disease heterogeneity and treatment response. The report concludes with ongoing gaps in our understanding of asthma pathobiology and provides some recommendations for future research to better understand the clinical and basic mechanisms underlying disease heterogeneity in asthma and to advance the development of new treatments for this growing public health problem.
Collapse
Affiliation(s)
- Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony N. Gerber
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York; and
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
7
|
Atochina-Vasserman E, Meshanni J, Stevenson E, Zhang D, Sun R, Ona N, Reagan E, Abramova E, Guo CJ, Wilkinson M, Baboo I, Yang Y, Pan L, Maurya D, Percec V, Li Y, Gow A, Weissman D. Targeted delivery of TGF-β mRNA to lung parenchyma using one-component ionizable amphiphilic Janus Dendrimers. RESEARCH SQUARE 2024:rs.3.rs-4656663. [PMID: 39041040 PMCID: PMC11261981 DOI: 10.21203/rs.3.rs-4656663/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Current clinical strategies for the delivery of pulmonary therapeutics to the lung are primarily targeted to the upper portions of the airways. However, targeted delivery to the lower regions of the lung is necessary for the treatment of parenchymal lung injury and disease. Here, we have developed an mRNA therapeutic for the lower lung using one-component Ionizable Amphiphilic Janus Dendrimers (IAJDs) as a delivery vehicle. We deliver an anti-inflammatory cytokine mRNA, transforming growth factor-beta (TGF-β), to produce transient protein expression in the lower regions of the lung. This study highlights IAJD's potential for precise, effective, and safe delivery of TGF-β mRNA to the lung. This delivery system offers a promising approach for targeting therapeutics to the specific tissues, a strategy necessary to fill the current clinical gap in treating parenchymal lung injury and disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Nathan Ona
- University of Pennsylvania Perelman School of Medicine
| | - Erin Reagan
- University of Pennsylvania Perelman School of Medicine
| | | | | | | | - Ishana Baboo
- University of Pennsylvania Perelman School of Medicine
| | - Yuzi Yang
- East China University of Science and Technology
| | - Liuyan Pan
- East China University of Science and Technology
| | - Devendra Maurya
- Roy & Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania
| | | | | | | | | |
Collapse
|
8
|
Hansen ME, Ibrahim Y, Desai TA, Koval M. Nanostructure-Mediated Transport of Therapeutics through Epithelial Barriers. Int J Mol Sci 2024; 25:7098. [PMID: 39000205 PMCID: PMC11241453 DOI: 10.3390/ijms25137098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The ability to precisely treat human disease is facilitated by the sophisticated design of pharmacologic agents. Nanotechnology has emerged as a valuable approach to creating vehicles that can specifically target organ systems, effectively traverse epithelial barriers, and protect agents from premature degradation. In this review, we discuss the molecular basis for epithelial barrier function, focusing on tight junctions, and describe different pathways that drugs can use to cross barrier-forming tissue, including the paracellular route and transcytosis. Unique features of drug delivery applied to different organ systems are addressed: transdermal, ocular, pulmonary, and oral delivery. We also discuss how design elements of different nanoscale systems, such as composition and nanostructured architecture, can be used to specifically enhance transepithelial delivery. The ability to tailor nanoscale drug delivery vehicles to leverage epithelial barrier biology is an emerging theme in the pursuit of facilitating the efficacious delivery of pharmacologic agents.
Collapse
Affiliation(s)
- M. Eva Hansen
- University of California Berkeley-University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yasmin Ibrahim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Graduate Program in Biochemistry, Cell and Developmental Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Tejal A. Desai
- University of California Berkeley-University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA;
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
9
|
Wei X, Liu N, Feng Y, Wang H, Han W, Zhuang M, Zhang H, Gao W, Lin Y, Tang X, Zheng Y. Competitive-like binding between carbon black and CTNNB1 to ΔNp63 interpreting the abnormal respiratory epithelial repair after injury. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172652. [PMID: 38653146 DOI: 10.1016/j.scitotenv.2024.172652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Airway epithelium is extraordinary vulnerable to damage owning to continuous environment exposure. Subsequent repair is therefore essential to restore the homeostasis of respiratory system. Disruptions in respiratory epithelial repair caused by nanoparticles exposure have been linked to various human diseases, yet implications in repair process remain incompletely elucidated. This study aims to elucidate the key stage in epithelial repair disturbed by carbon black (CB) nanoparticles, highlighting the pivotal role of ΔNp63 in mediating the epithelium repair. A competitive-like binding between CB and beta-catenin 1 (CTNNB1) to ΔNp63 is proposed to elaborate the underlying toxicity mechanism. Specifically, CB exhibits a remarkable inhibitory effect on cell proliferation, leading to aberrant airway epithelial repair, as validated in air-liquid culture. ΔNp63 drives efficient epithelial proliferation during CB exposure, and CTNNB1 was identified as a target of ΔNp63 by bioinformatics analysis. Further molecular dynamics simulation reveals that oxygen-containing functional groups on CB disrupt the native interaction of CTNNB1 with ΔNp63 through competitive-like binding pattern. This process modulates CTNNB1 expression, ultimately restraining proliferation during respiratory epithelial repair. Overall, the current study elucidates that the diminished interaction between CTNNB1 and ΔNp63 impedes respiratory epithelial repair in response to CB exposure, thereby enriching the public health risk assessment on CB-related respiratory diseases.
Collapse
Affiliation(s)
- Xiaoran Wei
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Nan Liu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yawen Feng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Hongmei Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Weizhong Han
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Min Zhuang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266003, China
| | - Hongna Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Wei Gao
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Tang
- Department of Medical Chemistry, School of Pharmacy, Qingdao University, Qingdao 266071, China.
| | - Yuxin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
10
|
Shahdab N, Ward C, Hansbro PM, Cummings S, Young JS, Moheimani F. Distinct Effects of Respiratory Viral Infection Models on miR-149-5p, IL-6 and p63 Expression in BEAS-2B and A549 Epithelial Cells. Cells 2024; 13:919. [PMID: 38891051 PMCID: PMC11172188 DOI: 10.3390/cells13110919] [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: 04/27/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Respiratory viruses cause airway inflammation, resulting in epithelial injury and repair. miRNAs, including miR-149-5p, regulate different pathological conditions. We aimed to determine how miR-149-5p functions in regulating pro-inflammatory IL-6 and p63, key regulators of airway epithelial wound repair, in response to viral proteins in bronchial (BEAS-2B) and alveolar (A549) epithelial cells. BEAS-2B or A549 cells were incubated with poly (I:C, 0.5 µg/mL) for 48 h or SARS-CoV-2 spike protein-1 or 2 subunit (S1 or S2, 1 μg/mL) for 24 h. miR-149-5p was suppressed in BEAS-2B challenged with poly (I:C), correlating with IL-6 and p63 upregulation. miR-149-5p was down-regulated in A549 stimulated with poly (I:C); IL-6 expression increased, but p63 protein levels were undetectable. miR-149-5p remained unchanged in cells exposed to S1 or S2, while S1 transfection increased IL-6 expression in BEAS-2B cells. Ectopic over-expression of miR-149-5p in BEAS-2B cells suppressed IL-6 and p63 mRNA levels and inhibited poly (I:C)-induced IL-6 and p63 mRNA expressions. miR-149-5p directly suppressed IL-6 mRNA in BEAS-2B cells. Hence, BEAS-2B cells respond differently to poly (I:C), S1 or S2 compared to A549 cells. Thus, miR-149-5p dysregulation may be involved in poly (I:C)-stimulated but not S1- or S2-stimulated increased IL-6 production and p63 expression in BEAS-2B cells.
Collapse
Affiliation(s)
- Nafeesa Shahdab
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Christopher Ward
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia;
| | - Stephen Cummings
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - John S. Young
- National Horizons Centre, School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK; (N.S.); (S.C.); (J.S.Y.)
| | - Fatemeh Moheimani
- Department of Life Sciences, Manchester Metropolitan University, Manchester M15 6BH, UK
| |
Collapse
|
11
|
Lee D, Hong JH. Chloride/Multiple Anion Exchanger SLC26A Family: Systemic Roles of SLC26A4 in Various Organs. Int J Mol Sci 2024; 25:4190. [PMID: 38673775 PMCID: PMC11050216 DOI: 10.3390/ijms25084190] [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] [Received: 03/01/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Solute carrier family 26 member 4 (SLC26A4) is a member of the SLC26A transporter family and is expressed in various tissues, including the airway epithelium, kidney, thyroid, and tumors. It transports various ions, including bicarbonate, chloride, iodine, and oxalate. As a multiple-ion transporter, SLC26A4 is involved in the maintenance of hearing function, renal function, blood pressure, and hormone and pH regulation. In this review, we have summarized the various functions of SLC26A4 in multiple tissues and organs. Moreover, the relationships between SLC26A4 and other channels, such as cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and sodium chloride cotransporter, are highlighted. Although the modulation of SLC26A4 is critical for recovery from malfunctions of various organs, development of specific inducers or agonists of SLC26A4 remains challenging. This review contributes to providing a better understanding of the role of SLC26A4 and development of therapeutic approaches for the SLC26A4-associated hearing loss and SLC26A4-related dysfunction of various organs.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, GAIHST (Gachon Advanced Institute for Health Sciences and Technology), Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
12
|
Bagley DC, Russell T, Ortiz-Zapater E, Stinson S, Fox K, Redd PF, Joseph M, Deering-Rice C, Reilly C, Parsons M, Brightling C, Rosenblatt J. Bronchoconstriction damages airway epithelia by crowding-induced excess cell extrusion. Science 2024; 384:66-73. [PMID: 38574138 DOI: 10.1126/science.adk2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/12/2024] [Indexed: 04/06/2024]
Abstract
Asthma is deemed an inflammatory disease, yet the defining diagnostic feature is mechanical bronchoconstriction. We previously discovered a conserved process called cell extrusion that drives homeostatic epithelial cell death when cells become too crowded. In this work, we show that the pathological crowding of a bronchoconstrictive attack causes so much epithelial cell extrusion that it damages the airways, resulting in inflammation and mucus secretion in both mice and humans. Although relaxing the airways with the rescue treatment albuterol did not affect these responses, inhibiting live cell extrusion signaling during bronchoconstriction prevented all these features. Our findings show that bronchoconstriction causes epithelial damage and inflammation by excess crowding-induced cell extrusion and suggest that blocking epithelial extrusion, instead of the ensuing downstream inflammation, could prevent the feed-forward asthma inflammatory cycle.
Collapse
Affiliation(s)
- Dustin C Bagley
- The Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King's College London, London SE1 1UL, UK
| | - Tobias Russell
- The Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King's College London, London SE1 1UL, UK
| | - Elena Ortiz-Zapater
- Department of Biochemistry and Molecular Biology, University of Valencia, 46010 Valencia, Spain
| | - Sally Stinson
- Institute for Lung Health, Leicester NIHR BRC, University of Leicester, Leicester LE3 9QP, UK
| | | | - Polly F Redd
- University of Utah, Salt Lake City, UT 84112, USA
| | - Merry Joseph
- University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | - Maddy Parsons
- The Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King's College London, London SE1 1UL, UK
| | - Christopher Brightling
- Institute for Lung Health, Leicester NIHR BRC, University of Leicester, Leicester LE3 9QP, UK
| | - Jody Rosenblatt
- The Randall Centre for Cell & Molecular Biophysics, School of Basic & Medical Biosciences, King's College London, London SE1 1UL, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London SE1 1UL, UK
| |
Collapse
|
13
|
Calzetta L, Page C, Matera MG, Cazzola M, Rogliani P. Use of human airway smooth muscle in vitro and ex vivo to investigate drugs for the treatment of chronic obstructive respiratory disorders. Br J Pharmacol 2024; 181:610-639. [PMID: 37859567 DOI: 10.1111/bph.16272] [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: 08/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023] Open
Abstract
Isolated airway smooth muscle has been extensively investigated since 1840 to understand the pharmacology of airway diseases. There has often been poor predictability from murine experiments to drugs evaluated in patients with asthma or chronic obstructive pulmonary disease (COPD). However, the use of isolated human airways represents a sensible strategy to optimise the development of innovative molecules for the treatment of respiratory diseases. This review aims to provide updated evidence on the current uses of isolated human airways in validated in vitro methods to investigate drugs in development for the treatment of chronic obstructive respiratory disorders. This review also provides historical notes on the pioneering pharmacological research on isolated human airway tissues, the key differences between human and animal airways, as well as the pivotal differences between human medium bronchi and small airways. Experiments carried out with isolated human bronchial tissues in vitro and ex vivo replicate many of the main anatomical, pathophysiological, mechanical and immunological characteristics of patients with asthma or COPD. In vitro models of asthma and COPD using isolated human airways can provide information that is directly translatable into humans with obstructive lung diseases. Regardless of the technique used to investigate drugs for the treatment of chronic obstructive respiratory disorders (i.e., isolated organ bath systems, videomicroscopy and wire myography), the most limiting factors to produce high-quality and repeatable data remain closely tied to the manual skills of the researcher conducting experiments and the availability of suitable tissue.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Clive Page
- Pulmonary Pharmacology Unit, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
14
|
Peña-García PE, Fastiggi VA, Mank MM, Ather JL, Garrow OJ, Anathy V, Dixon AE, Poynter ME. Bariatric surgery decreases the capacity of plasma from obese asthmatic subjects to augment airway epithelial cell proinflammatory cytokine production. Am J Physiol Lung Cell Mol Physiol 2024; 326:L71-L82. [PMID: 37988602 PMCID: PMC11292671 DOI: 10.1152/ajplung.00205.2023] [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] [Received: 07/02/2023] [Revised: 11/03/2023] [Accepted: 11/18/2023] [Indexed: 11/23/2023] Open
Abstract
Obesity is a risk factor for asthma. Individuals with asthma and obesity often have poor asthma control and do not respond as well to therapies such as inhaled corticosteroids and long-acting bronchodilators. Weight loss improves asthma control, with a 5%-10% loss in body mass necessary and sufficient to lead to clinically relevant improvements. Preclinical studies have demonstrated the pathogenic contribution of adipocytes from obese mice to the augmented production of proinflammatory cytokines from airway epithelial cells and the salutary effects of diet-induced weight loss to decrease these consequences. However, the effects of adipocyte-derived products on airway epithelial function in human obesity remain incompletely understood. We utilized samples collected from a 12-mo longitudinal study of subjects with obesity undergoing weight loss (bariatric) surgery including controls without asthma and subjects with allergic and nonallergic obese asthma. Visceral adipose tissue (VAT) samples were collected during bariatric surgery and from recruited normal weight controls without asthma undergoing elective abdominal surgery. Human bronchial epithelial (HBEC3-KT) cells were exposed to plasma or conditioned media from cultured VAT adipocytes with or without agonists. Human bronchial smooth muscle (HBSM) cells were similarly exposed to adipocyte-conditioned media. Proinflammatory cytokines were augmented in supernatants from HBEC3-KT cells exposed to plasma as compared with subsequent visits. Whereas exposure to obese adipocyte-conditioned media induced proinflammatory responses, there were no differences between groups in both HBEC3-KT and HBSM cells. These data show that bariatric surgery and subsequent weight loss beneficially change the circulating factors that augment human airway epithelial and bronchial smooth muscle cell proinflammatory responses.NEW & NOTEWORTHY This longitudinal study following subjects with asthma and obesity reveals that weight loss following bariatric surgery decreases the capacity for plasma to augment proinflammatory cytokine secretion by human bronchial epithelial cells, implicating that circulating but not adipocyte-derived factors are important modulators in obese asthma.
Collapse
Affiliation(s)
- Paola E Peña-García
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
- Cellular, Molecular, and Biomedical Sciences doctoral program, University of Vermont, Burlington, Vermont, United States
| | - V Amanda Fastiggi
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
- Cellular, Molecular, and Biomedical Sciences doctoral program, University of Vermont, Burlington, Vermont, United States
| | - Madeleine M Mank
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
| | - Jennifer L Ather
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
| | - Olivia J Garrow
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
| | - Vikas Anathy
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, United States
| | - Anne E Dixon
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
| | - Matthew E Poynter
- Vermont Lung Center, University of Vermont, Burlington, Vermont, United States
- Pulmonary Disease and Critical Care Medicine, University of Vermont, Burlington, Vermont, United States
| |
Collapse
|
15
|
Bansal A, Kooi C, Kalyanaraman K, Gill S, Thorne A, Chandramohan P, Necker-Brown A, Mostafa MM, Milani A, Leigh R, Newton R. Synergy between Interleukin-1 β, Interferon- γ, and Glucocorticoids to Induce TLR2 Expression Involves NF- κB, STAT1, and the Glucocorticoid Receptor. Mol Pharmacol 2023; 105:23-38. [PMID: 37863662 DOI: 10.1124/molpharm.123.000740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/22/2023] Open
Abstract
Glucocorticoids act via the glucocorticoid receptor (GR; NR3C1) to downregulate inflammatory gene expression and are effective treatments for mild to moderate asthma. However, in severe asthma and virus-induced exacerbations, glucocorticoid therapies are less efficacious, possibly due to reduced repressive ability and/or the increased expression of proinflammatory genes. In human A549 epithelial and primary human bronchial epithelial cells, toll-like receptor (TLR)-2 mRNA and protein were supra-additively induced by interleukin-1β (IL-1β) plus dexamethasone (IL-1β+Dex), interferon-γ (IFN-γ) plus dexamethasone (IFN-γ+Dex), and IL-1β plus IFN-γ plus dexamethasone (IL-1β+IFN-γ+Dex). Indeed, ∼34- to 2100-fold increases were apparent at 24 hours for IL-1β+IFN-γ+Dex, and this was greater than for any single or dual treatment. Using the A549 cell model, TLR2 induction by IL-1β+IFN-γ+Dex was antagonized by Org34517, a competitive GR antagonist. Further, when combined with IL-1β, IFN-γ, or IL-1β+IFN-γ, the enhancements by dexamethasone on TLR2 expression required GR. Likewise, inhibitor of κB kinase 2 inhibitors reduced IL-1β+IFN-γ+Dex-induced TLR2 expression, and TLR2 expression induced by IL-1β+Dex, with or without IFN-γ, required the nuclear factor (NF)-κB subunit, p65. Similarly, signal transducer and activator of transcription (STAT)-1 phosphorylation and γ-interferon-activated sequence-dependent transcription were induced by IFN-γ These, along with IL-1β+IFN-γ+Dex-induced TLR2 expression, were inhibited by Janus kinase (JAK) inhibitors. As IL-1β+IFN-γ+Dex-induced TLR2 expression also required STAT1, this study reveals cooperation between JAK-STAT1, NF-κB, and GR to upregulate TLR2 expression. Since TLR2 agonism elicits inflammatory responses, we propose that synergies involving TLR2 may occur within the cytokine milieu present in the immunopathology of glucocorticoid-resistant disease, and this could promote glucocorticoid resistance. SIGNIFICANCE STATEMENT: This study highlights that in human pulmonary epithelial cells, glucocorticoids, when combined with the inflammatory cytokines interleukin-1β (IL-1β) and interferon-γ (IFN-γ), can synergistically induce the expression of inflammatory genes, such as TLR2. This effect involved positive combinatorial interactions between NF-κB/p65, glucocorticoid receptor, and JAK-STAT1 signaling to synergistically upregulate TLR2 expression. Thus, synergies involving glucocorticoid enhancement of TLR2 expression may occur in the immunopathology of glucocorticoid-resistant inflammatory diseases, including severe asthma.
Collapse
Affiliation(s)
- Akanksha Bansal
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Cora Kooi
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Keerthana Kalyanaraman
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Sachman Gill
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Andrew Thorne
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Priyanka Chandramohan
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Amandah Necker-Brown
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Mahmoud M Mostafa
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Arya Milani
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Richard Leigh
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Robert Newton
- Departments of Physiology and Pharmacology (A.B., K.K., S.G., A.T., P.C., A.N.-B., M.M.M., A.M., R.N.) and Medicine (C.K., R.L.), Lung Health Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, Canada
| |
Collapse
|
16
|
Guo TJF, Singhera GK, Leung JM, Dorscheid DR. Airway Epithelial-Derived Immune Mediators in COVID-19. Viruses 2023; 15:1655. [PMID: 37631998 PMCID: PMC10458661 DOI: 10.3390/v15081655] [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] [Received: 06/13/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Tony J. F. Guo
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
17
|
Leach T, Gandhi U, Reeves KD, Stumpf K, Okuda K, Marini FC, Walker SJ, Boucher R, Chan J, Cox LA, Atala A, Murphy SV. Development of a novel air-liquid interface airway tissue equivalent model for in vitro respiratory modeling studies. Sci Rep 2023; 13:10137. [PMID: 37349353 PMCID: PMC10287689 DOI: 10.1038/s41598-023-36863-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
The human airways are complex structures with important interactions between cells, extracellular matrix (ECM) proteins and the biomechanical microenvironment. A robust, well-differentiated in vitro culture system that accurately models these interactions would provide a useful tool for studying normal and pathological airway biology. Here, we report the development and characterization of a physiologically relevant air-liquid interface (ALI) 3D airway 'organ tissue equivalent' (OTE) model with three novel features: native pulmonary fibroblasts, solubilized lung ECM, and hydrogel substrate with tunable stiffness and porosity. We demonstrate the versatility of the OTE model by evaluating the impact of these features on human bronchial epithelial (HBE) cell phenotype. Variations of this model were analyzed during 28 days of ALI culture by evaluating epithelial confluence, trans-epithelial electrical resistance, and epithelial phenotype via multispectral immuno-histochemistry and next-generation sequencing. Cultures that included both solubilized lung ECM and native pulmonary fibroblasts within the hydrogel substrate formed well-differentiated ALI cultures that maintained a barrier function and expressed mature epithelial markers relating to goblet, club, and ciliated cells. Modulation of hydrogel stiffness did not negatively impact HBE differentiation and could be a valuable variable to alter epithelial phenotype. This study highlights the feasibility and versatility of a 3D airway OTE model to model the multiple components of the human airway 3D microenvironment.
Collapse
Affiliation(s)
- Timothy Leach
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
- Wake Forest School of Medicine, Medical Center Boulevard, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Winston-Salem, NC, 27157, USA
| | - Uma Gandhi
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Kimberly D Reeves
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kristina Stumpf
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Kenichi Okuda
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Frank C Marini
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Stephen J Walker
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
| | - Richard Boucher
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jeannie Chan
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Laura A Cox
- Center for Precision Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Anthony Atala
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA
- Wake Forest School of Medicine, Medical Center Boulevard, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Winston-Salem, NC, 27157, USA
| | - Sean V Murphy
- Wake Forest School of Medicine, Medical Center, Wake Forest Institute for Regenerative Medicine, 391 Technology Way, Winston-Salem, NC, 27101, USA.
- Wake Forest School of Medicine, Medical Center Boulevard, Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
18
|
Barbosa-de-Oliveira MC, Oliveira-Melo P, Gonçalves da Silva MH, Santos da Silva F, Andrade Carvalho da Silva F, Silva de Araujo BV, Franco de Oliveira M, Tadeu Correia A, Miyoshi Sakamoto S, Valença SS, Lanzetti M, Schmidt M, Kennedy-Feitosa E. Modulation of Alveolar Macrophage Activity by Eugenol Attenuates Cigarette-Smoke-Induced Acute Lung Injury in Mice. Antioxidants (Basel) 2023; 12:1258. [PMID: 37371988 DOI: 10.3390/antiox12061258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
This study investigates the role of eugenol (EUG) on CS-induced acute lung injury (ALI) and how this compound is able to modulate macrophage activity. C57BL/6 mice were exposed to 12 cigarettes/day/5days and treated 15 min/day/5days with EUG. Rat alveolar macrophages (RAMs) were exposed to CSE (5%) and treated with EUG. In vivo, EUG reduced morphological changes inflammatory cells, oxidative stress markers, while, in vitro, it induced balance in the oxidative stress and reduced the pro-inflammatory cytokine release while increasing the anti-inflammatory one. These results suggest that eugenol reduced CS-induced ALI and acted as a modulator of macrophage activity.
Collapse
Affiliation(s)
- Maria Clara Barbosa-de-Oliveira
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| | - Paolo Oliveira-Melo
- Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Universidade de Sao Paulo, Sao Paulo 05508-220, Brazil
| | | | - Flávio Santos da Silva
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| | - Felipe Andrade Carvalho da Silva
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| | - Bruno Vinicios Silva de Araujo
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| | | | - Aristides Tadeu Correia
- Laboratório de Pesquisa em Cirurgia Torácica, Faculdade de Medicina HCFMUSP, Universidade de Sao Paulo, Sao Paulo 05508-220, Brazil
| | - Sidnei Miyoshi Sakamoto
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| | - Samuel Santos Valença
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil
| | - Manuella Lanzetti
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-590, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, Building 3211, Room 406, 9713 AV Groningen, The Netherlands
- Groningen Research Institute of Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Emanuel Kennedy-Feitosa
- Morphophysiopharmacology Laboratory, Department of Health Sciences, Federal University of the Semi-Arid Region, Mossoró 59625-900, Brazil
| |
Collapse
|
19
|
Zhu Y, Chang D. Interactions between the lung microbiome and host immunity in chronic obstructive pulmonary disease. Chronic Dis Transl Med 2023; 9:104-121. [PMID: 37305112 PMCID: PMC10249200 DOI: 10.1002/cdt3.66] [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: 02/02/2023] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease and the third leading cause of death worldwide. Developments in next-generation sequencing technology have improved microbiome analysis, which is increasingly recognized as an important component of disease management. Similar to the gut, the lung is a biosphere containing billions of microbial communities. The lung microbiome plays an important role in regulating and maintaining the host immune system. The microbiome composition, metabolites of microorganisms, and the interactions between the lung microbiome and the host immunity profoundly affect the occurrence, development, treatment, and prognosis of COPD. In this review, we drew comparisons between the lung microbiome of healthy individuals and that of patients with COPD. Furthermore, we summarize the intrinsic interactions between the host and the overall lung microbiome, focusing on the underlying mechanisms linking the microbiome to the host innate and adaptive immune response pathways. Finally, we discuss the possibility of using the microbiome as a biomarker to determine the stage and prognosis of COPD and the feasibility of developing a novel, safe, and effective therapeutic target.
Collapse
Affiliation(s)
- Yixing Zhu
- Graduate School of The PLA General HospitalBeijingChina
| | - De Chang
- Department of Respiratory and Critical Care Medicine, Eighth Medical Center, Department of Respiratory and Critical Care Seventh Medical CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
20
|
TSLP and HMGB1: Inflammatory Targets and Potential Biomarkers for Precision Medicine in Asthma and COPD. Biomedicines 2023; 11:biomedicines11020437. [PMID: 36830972 PMCID: PMC9953666 DOI: 10.3390/biomedicines11020437] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The airway epithelium, through pattern recognition receptors expressed transmembrane or intracellularly, acts as a first line of defense for the lungs against many environmental triggers. It is involved in the release of alarmin cytokines, which are important mediators of inflammation, with receptors widely expressed in structural cells as well as innate and adaptive immune cells. Knowledge of the role of epithelial cells in orchestrating the immune response and mediating the clearance of invading pathogens and dead/damaged cells to facilitate resolution of inflammation is necessary to understand how, in many chronic lung diseases, there is a persistent inflammatory response that becomes the basis of underlying pathogenesis. This review will focus on the role of pulmonary epithelial cells and of airway epithelial cell alarmins, in particular thymic stromal lymphopoietin (TSLP) and high mobility group box 1 (HMGB1), as key mediators in driving the inflammation of chronic lung diseases, such as asthma and chronic obstructive pulmonary disease (COPD), evaluating the similarities and differences. Moreover, emerging concepts regarding the therapeutic role of molecules that act on airway epithelial cell alarmins will be explored for a precision medicine approach in the context of pulmonary diseases, thus allowing the use of these molecules as possible predictive biomarkers of clinical and biological response.
Collapse
|
21
|
Vitamin A-regulated ciliated cells promote airway epithelium repair in an asthma mouse model. Allergol Immunopathol (Madr) 2023; 51:116-125. [PMID: 36617830 DOI: 10.15586/aei.v51i1.700] [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: 06/14/2022] [Accepted: 10/13/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Asthma is a chronic inflammatory airway disease that causes damage to and exfoliation of the airway epithelium. The continuous damage to the airway epithelium in asthma cannot be repaired quickly and generates irreversible damage, repeated attacks, and aggravation. Vitamin A (VA) has multifarious biological functions that include maintaining membrane stability and integrity of the structure and function of epithelial cells. Our research explored the role of VA in repairing the airway epithelium and provided a novel treatment strategy for asthma. METHODS A mouse asthma model was established by house dust mite (HDM) and treated with VA by gavage. Human bronchial epithelial (16HBE) cells were treated with HDM and all-trans retinoic acid (ATRA) in vitro. We analyzed the mRNA and protein expression of characteristic markers, such as acetyl-α-tubulin (Ac-TUB) and FOXJ1 in ciliated cells and MUC5AC in secretory cells, mucus secretion, airway inflammation, the morphology of cilia, and the integrity of the airway epithelium. RESULTS Findings showed destruction of airway epithelial integrity, damaged cilia, high mucus secretion, increased MUC5AC expression, and decreased Ac-TUB and FOXJ1 expression in asthmatic mice. The VA intervention reversed the effect on Ac-TUB and FOXJ1 and promoted ciliated cells to repair the damage and maintain airway epithelial integrity. In 16HBE cells, we could confirm that ATRA promoted the expression of Ac-TUB and FOXJ1. CONCLUSION These results demonstrated that VA-regulated ciliated cells to repair the damaged airway epithelium caused by asthma and maintain airway epithelial integrity. VA intervention is a potential adjunct to conventional treatment for asthma.
Collapse
|
22
|
Ni K, Che B, Yang C, Qin Y, Gu R, Wang C, Luo M, Deng L. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Front Pharmacol 2022; 13:1033043. [PMID: 36578545 PMCID: PMC9790924 DOI: 10.3389/fphar.2022.1033043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a never before seen challenge to human health and the world economy. However, it is difficult to widely use conventional animal and cell culture models in understanding the underlying pathological mechanisms of COVID-19, which in turn hinders the development of relevant therapeutic treatments, including drugs. To overcome this challenge, various three-dimensional (3D) pulmonary cell culture models such as organoids are emerging as an innovative toolset for simulating the pathophysiology occurring in the respiratory system, including bronchial airways, alveoli, capillary network, and pulmonary interstitium, which provide a robust and powerful platform for studying the process and underlying mechanisms of SARS-CoV-2 infection among the potential primary targets in the lung. This review introduces the key features of some of these recently developed tools, including organoid, lung-on-a-chip, and 3D bioprinting, which can recapitulate different structural compartments of the lung and lung function, in particular, accurately resembling the human-relevant pathophysiology of SARS-CoV-2 infection in vivo. In addition, the recent progress in developing organoids for alveolar and airway disease modeling and their applications for discovering drugs against SARS-CoV-2 infection are highlighted. These innovative 3D cell culture models together may hold the promise to fully understand the pathogenesis and eventually eradicate the pandemic of COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| |
Collapse
|
23
|
Zhong B, Seah JJ, Liu F, Ba L, Du J, Wang DY. The role of hypoxia in the pathophysiology of chronic rhinosinusitis. Allergy 2022; 77:3217-3232. [PMID: 35603933 DOI: 10.1111/all.15384] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/19/2022] [Accepted: 05/17/2022] [Indexed: 02/05/2023]
Abstract
Chronic rhinosinusitis (CRS) is a chronic inflammatory disease of the nasal cavity characterized by excessive nasal mucus secretion and nasal congestion. The development of CRS is related to pathological mechanisms induced by hypoxia. Under hypoxic conditions, the stable expression of both Hypoxia inducible factor-1 (HIF-1) α and HIF-2α are involved in the immune response and inflammatory pathways of CRS. The imbalance in the composition of nasal microbiota may affect the hypoxic state of CRS and perpetuate existing inflammation. Hypoxia affects the differentiation of nasal epithelial cells such as ciliated cells and goblet cells, induces fibroblast proliferation, and leads to epithelial-mesenchymal transition (EMT) and tissue remodeling. Hypoxia also affects the proliferation and differentiation of macrophages, eosinophils, basophils, and mast cells in sinonasal mucosa, and thus influences the inflammatory state of CRS by regulating T cells and B cells. Given the multifactorial nature in which HIF is linked to CRS, this study aims to elucidate the effect of hypoxia on the pathogenic mechanisms of CRS.
Collapse
Affiliation(s)
- Bing Zhong
- Upper Airways Research Laboratory, Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China.,Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jun Jie Seah
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Feng Liu
- Upper Airways Research Laboratory, Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Luo Ba
- Department of Otolaryngology, People's Hospital of Tibet Autonomous Region, Lhasa, China
| | - Jintao Du
- Upper Airways Research Laboratory, Department of Otolaryngology-Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - De Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Ortiz-Zapater E, Bagley DC, Hernandez VL, Roberts LB, Maguire TJA, Voss F, Mertins P, Kirchner M, Peset-Martin I, Woszczek G, Rosenblatt J, Gotthardt M, Santis G, Parsons M. Epithelial coxsackievirus adenovirus receptor promotes house dust mite-induced lung inflammation. Nat Commun 2022; 13:6407. [PMID: 36302767 PMCID: PMC9613683 DOI: 10.1038/s41467-022-33882-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 10/06/2022] [Indexed: 12/25/2022] Open
Abstract
Airway inflammation and remodelling are important pathophysiologic features in asthma and other respiratory conditions. An intact epithelial cell layer is crucial to maintain lung homoeostasis, and this depends on intercellular adhesion, whilst damaged respiratory epithelium is the primary instigator of airway inflammation. The Coxsackievirus Adenovirus Receptor (CAR) is highly expressed in the epithelium where it modulates cell-cell adhesion stability and facilitates immune cell transepithelial migration. However, the contribution of CAR to lung inflammation remains unclear. Here we investigate the mechanistic contribution of CAR in mediating responses to the common aeroallergen, House Dust Mite (HDM). We demonstrate that administration of HDM in mice lacking CAR in the respiratory epithelium leads to loss of peri-bronchial inflammatory cell infiltration, fewer goblet-cells and decreased pro-inflammatory cytokine release. In vitro analysis in human lung epithelial cells confirms that loss of CAR leads to reduced HDM-dependent inflammatory cytokine release and neutrophil migration. Epithelial CAR depletion also promoted smooth muscle cell proliferation mediated by GSK3β and TGF-β, basal matrix production and airway hyperresponsiveness. Our data demonstrate that CAR coordinates lung inflammation through a dual function in leucocyte recruitment and tissue remodelling and may represent an important target for future therapeutic development in inflammatory lung diseases.
Collapse
Affiliation(s)
- Elena Ortiz-Zapater
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
| | - Dustin C Bagley
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | | | - Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Thomas J A Maguire
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Felizia Voss
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- DZHK Partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Marieluise Kirchner
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | | - Grzegorz Woszczek
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jody Rosenblatt
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Michael Gotthardt
- Max-Delbrück-Centrum für Molekulare Medizin in the Helmholtz Assoziation (MDC), Berlin, Germany
- Berlin Institute of Health at Charité, Universitaetsmedizin Berlin, Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Charité Universitätsmedizin Berlin, Berlin, Germany
| | - George Santis
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Science King's College London, London, UK
- Department of Respiratory Medicine, Guy's & St Thomas NHS Trust, London, UK
| | - Maddy Parsons
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| |
Collapse
|
25
|
Stevenson ER, Wilkinson ML, Abramova E, Guo C, Gow AJ. Intratracheal Administration of Acyl Coenzyme A Acyltransferase-1 Inhibitor K-604 Reduces Pulmonary Inflammation Following Bleomycin-Induced Lung Injury. J Pharmacol Exp Ther 2022; 382:356-365. [PMID: 35970601 PMCID: PMC9426763 DOI: 10.1124/jpet.122.001284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 01/19/2023] Open
Abstract
Acute lung injury (ALI) is characterized by epithelial damage, barrier dysfunction, and pulmonary edema. Macrophage activation and failure to resolve play a role in ALI; thus, macrophage phenotype modulation is a rational target for therapeutic intervention. Large, lipid-laden macrophages have been observed in various injury models, including intratracheal bleomycin (ITB), suggesting that lipid storage may play a role in ALI severity. The endoplasmic reticulum-associated enzyme acyl coenzyme A acyltransferase-1 (Acat-1/Soat1) is highly expressed in macrophages, where it catalyzes the esterification of cholesterol, leading to intracellular lipid accumulation. We hypothesize that inhibition of Acat-1 will reduce macrophage activation and improve outcomes of lung injury in ITB. K-604, a selective inhibitor of Acat-1, was used to reduce cholesterol esterification and hence lipid accumulation in response to ITB. Male and female C57BL6/J mice (n = 16-21/group) were administered control, control + K-604, ITB, or ITB + K-604 on d0, control or K-604 on d3, and were sacrificed on day 7. ITB caused significant body weight loss and an increase in cholesterol accumulation in bronchoalveolar lavage cells. These changes were mitigated by Acat-1 inhibition. K-604 also significantly reduced ITB-induced alveolar thickening. Surfactant composition was normalized as indicated by a significant decrease in phospholipid: SP-B ratio in ITB+K-604 compared with ITB. K-604 administration preserved mature alveolar macrophages, decreased activation in response to ITB, and decreased the percentage mature and pro-fibrotic interstitial macrophages. These results show that inhibition of Acat-1 in the lung is associated with reduced inflammatory response to ITB-mediated lung injury. SIGNIFICANCE STATEMENT: Acyl coenzyme A acyltransferase-1 (Acat-1) is critical to lipid droplet formation, and thus inhibition of Acat-1 presents as a pharmacological target. Intratracheal administration of K-604, an Acat-1 inhibitor, reduces intracellular cholesterol ester accumulation in lung macrophages, attenuates inflammation and macrophage activation, and normalizes mediators of surface-active function after intratracheal bleomycin administration in a rodent model. The data presented within suggest that inhibition of Acat-1 in the lung improves acute lung injury outcomes.
Collapse
Affiliation(s)
- Emily R Stevenson
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Melissa L Wilkinson
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Elena Abramova
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Changjiang Guo
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Andrew J Gow
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
26
|
Gandhi VD, Shrestha Palikhe N, Vliagoftis H. Protease-activated receptor-2: Role in asthma pathogenesis and utility as a biomarker of disease severity. Front Med (Lausanne) 2022; 9:954990. [PMID: 35966869 PMCID: PMC9372307 DOI: 10.3389/fmed.2022.954990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
PAR2, a receptor activated by serine proteases, has primarily pro-inflammatory roles in the airways and may play a role in asthma pathogenesis. PAR2 exerts its effects in the lungs through activation of a variety of airway cells, but also activation of circulating immune cells. There is evidence that PAR2 expression increases in asthma and other inflammatory diseases, although the regulation of PAR2 expression is not fully understood. Here we review the available literature on the potential role of PAR2 in asthma pathogenesis and propose a model of PAR2-mediated development of allergic sensitization. We also propose, based on our previous work, that PAR2 expression on peripheral blood monocyte subsets has the potential to serve as a biomarker of asthma severity and/or control.
Collapse
Affiliation(s)
- Vivek Dipak Gandhi
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Nami Shrestha Palikhe
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Alberta Respiratory Centre, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Harissios Vliagoftis,
| |
Collapse
|
27
|
Qiu ZE, Xu JB, Chen L, Huang ZX, Lei TL, Huang ZY, Hou XC, Yang HL, Lin QH, Zhu YX, Zhao L, Zhou WL, Zhang YL. Allicin Facilitates Airway Surface Liquid Hydration by Activation of CFTR. Front Pharmacol 2022; 13:890284. [PMID: 35784719 PMCID: PMC9241074 DOI: 10.3389/fphar.2022.890284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
Airway epithelium plays critical roles in regulating airway surface liquid (ASL), the alteration of which causes mucus stasis symptoms. Allicin is a compound released from garlic and harbors the capacity of lung-protection. However, the potential regulatory effects of allicin on airway epithelium remain elusive. This study aimed to investigate the effects of allicin on ion transport across airway epithelium and evaluate its potential as an expectorant. Application of allicin induced Cl− secretion across airway epithelium in a concentration-dependent manner. Blockade of cystic fibrosis transmembrane conductance regulator (CFTR) or inhibition of adenylate cyclase-cAMP signaling pathway attenuated allicin-induced Cl− secretion in airway epithelial cells. The in vivo study showed that inhaled allicin significantly increased the ASL secretion in mice. These results suggest that allicin induces Cl− and fluid secretion across airway epithelium via activation of CFTR, which might provide therapeutic strategies for the treatment of chronic pulmonary diseases associated with ASL dehydration.
Collapse
Affiliation(s)
- Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian-Bang Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Tian-Lun Lei
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zi-Yang Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hai-Long Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qin-Hua Lin
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Lei Zhao, ; Wen-Liang Zhou, ; Yi-Lin Zhang,
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Lei Zhao, ; Wen-Liang Zhou, ; Yi-Lin Zhang,
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Lei Zhao, ; Wen-Liang Zhou, ; Yi-Lin Zhang,
| |
Collapse
|
28
|
Aikaterini G, Tomás S, Ilias K, Christina R, Yu-Wei L, Mina P, Spyros Z, Helen G, Jian L, E FL. Pulmonary and systemic pharmacokinetics of colistin methanesulfonate (CMS) and formed colistin following nebulization of CMS among patients with ventilator-associated pneumonia. Int J Antimicrob Agents 2022; 59:106588. [DOI: 10.1016/j.ijantimicag.2022.106588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 02/05/2022] [Accepted: 04/03/2022] [Indexed: 11/16/2022]
|
29
|
Albano GD, Montalbano AM, Gagliardo R, Anzalone G, Profita M. Impact of Air Pollution in Airway Diseases: Role of the Epithelial Cells (Cell Models and Biomarkers). Int J Mol Sci 2022; 23:2799. [PMID: 35269941 PMCID: PMC8911203 DOI: 10.3390/ijms23052799] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 02/05/2023] Open
Abstract
Biomedical research is multidisciplinary and often uses integrated approaches performing different experimental models with complementary functions. This approach is important to understand the pathogenetic mechanisms concerning the effects of environmental pollution on human health. The biological activity of the substances is investigated at least to three levels using molecular, cellular, and human tissue models. Each of these is able to give specific answers to experimental problems. A scientific approach, using biological methods (wet lab), cell cultures (cell lines or primary), isolated organs (three-dimensional cell cultures of primary epithelial cells), and animal organisms, including the human body, aimed to understand the effects of air pollution on the onset of diseases of the respiratory system. Biological methods are divided into three complementary models: in vitro, ex vivo, and in vivo. In vitro experiments do not require the use of whole organisms (in vivo study), while ex vivo experiments use isolated organs or parts of organs. The concept of complementarity and the informatic support are useful tools to organize, analyze, and interpret experimental data, with the aim of discussing scientific notions with objectivity and rationality in biology and medicine. In this scenario, the integrated and complementary use of different experimental models is important to obtain useful and global information that allows us to identify the effect of inhaled pollutants on the incidence of respiratory diseases in the exposed population. In this review, we focused our attention on the impact of air pollution in airway diseases with a rapid and descriptive analysis on the role of epithelium and on the experimental cell models useful to study the effect of toxicants on epithelial cells.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Angela Marina Montalbano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Rosalia Gagliardo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Giulia Anzalone
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Mirella Profita
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| |
Collapse
|
30
|
Gadalla D, Tchoukalova YD, Lott DG. Regenerating airway epithelium using fibrous biomimetic basement membranes. J Biomed Mater Res A 2022; 110:1251-1262. [PMID: 35142434 DOI: 10.1002/jbm.a.37371] [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: 08/04/2021] [Revised: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/09/2022]
Abstract
There are reciprocal interactions between epithelial cells and underlying basement membrane. The resemblance of biomaterials to native basement membrane is thus critical for their success when used to regenerate epithelium-containing organs. Particularly, the use of nanofibers and the incorporation of basement membrane proteins may mimic both biophysical and biochemical properties of basement membrane, respectively. Herein we tested how electrospun polycaprolactone/heparin fibers with and without adsorbed laminin and collagen IV proteins affect epithelial cell functions. We found that airway epithelial cells attached, migrated, and proliferated on all scaffolds but protein-functionalized fibers promoted higher attachment, quicker migration, and increased proliferation. Fibers were then integrated on polyethylene scaffolds and cultured at an air-liquid interface. The detection of secretory and ciliated cell markers was higher in cells on polyethylene with fibers. These findings demonstrate that electrospun fibers incite beneficial epithelial cell responses and can be used in the fabrication of bioengineered functional epithelia.
Collapse
Affiliation(s)
- Dina Gadalla
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Yourka D Tchoukalova
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - David G Lott
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Phoenix, Arizona, USA.,Division of Laryngology, Mayo Clinic Arizona, Phoenix, Arizona, USA
| |
Collapse
|
31
|
Reconstituted basement membrane enables airway epithelium modeling and nanoparticle toxicity testing. Int J Biol Macromol 2022; 204:300-309. [PMID: 35149090 DOI: 10.1016/j.ijbiomac.2022.02.018] [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: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/14/2022]
Abstract
Basement membrane (BM) acts as a sheet-like extracellular matrix to support and promote the formation of epithelial and endothelial cell layers. The in vitro reconstruction of the BM is however not easy due to its ultrathin membrane features. This difficulty is overcome by self-assembling type IV collagen and laminin in the porous areas of a monolayer of crosslinked gelatin nanofibers deposited on a honeycomb microframe. Herein, a method is presented to generate airway epithelium by using such an artificial basement membrane (ABM) and human-induced pluripotent stem cells (hiPSCs). Bipolar primordial lung progenitors are firstly induced from hiPSCs and then replated on the ABM for differentiation toward matured airway epithelium under submerged and air-liquid interface culture conditions. As a result, a pseudostratified airway epithelium consisting of several cell types is achieved, showing remarkable apical secretion of MUC5AC proteins and clear advantages over other types of substrates. As a proof of concept, the derived epithelium is used for toxicity test of cadmium telluride (CdTe) nanoparticles (NPs), demonstrating the applicability of ABM-based assays involving hiPSC-derived epithelial cells-based assays.
Collapse
|
32
|
Morgan R, Manfredi C, Easley KF, Watkins LD, Hunt WR, Goudy SL, Sorscher EJ, Koval M, Molina SA. A medium composition containing normal resting glucose that supports differentiation of primary human airway cells. Sci Rep 2022; 12:1540. [PMID: 35087167 PMCID: PMC8795386 DOI: 10.1038/s41598-022-05446-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
Primary cells isolated from the human respiratory tract are the state-of-the-art for in vitro airway epithelial cell research. Airway cell isolates require media that support expansion of cells in a basal state to maintain the capacity for differentiation as well as proper cellular function. By contrast, airway cell differentiation at an air-liquid interface (ALI) requires a distinct medium formulation that typically contains high levels of glucose. Here, we expanded and differentiated human basal cells isolated from the nasal and conducting airway to a mature mucociliary epithelial cell layer at ALI using a medium formulation containing normal resting glucose levels. Of note, bronchial epithelial cells expanded and differentiated in normal resting glucose medium showed insulin-stimulated glucose uptake which was inhibited by high glucose concentrations. Normal glucose containing ALI also enabled differentiation of nasal and tracheal cells that showed comparable electrophysiological profiles when assessed for cystic fibrosis transmembrane conductance regulator (CFTR) function and that remained responsive for up to 7 weeks in culture. These data demonstrate that normal glucose containing medium supports differentiation of primary nasal and lung epithelial cells at ALI, is well suited for metabolic studies, and avoids pitfalls associated with exposure to high glucose.
Collapse
Affiliation(s)
- Rachel Morgan
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Candela Manfredi
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kristen F Easley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Lionel D Watkins
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - William R Hunt
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Steven L Goudy
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric J Sorscher
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Michael Koval
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Samuel A Molina
- Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, 205 Whitehead Building, 615 Michael Street, Atlanta, GA, 30322, USA
| |
Collapse
|
33
|
Sunil AA, Skaria T. Novel regulators of airway epithelial barrier function during inflammation: potential targets for drug repurposing. Expert Opin Ther Targets 2022; 26:119-132. [PMID: 35085478 DOI: 10.1080/14728222.2022.2035720] [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: 11/04/2022]
Abstract
INTRODUCTION Endogenous inflammatory signaling molecules resulting from deregulated immune responses, can impair airway epithelial barrier function and predispose individuals with airway inflammatory diseases to exacerbations and lung infections. Targeting the specific endogenous factors disrupting the airway barrier therefore has the potential to prevent disease exacerbations without affecting the protective immune responses. AREAS COVERED Here, we review the endogenous factors and specific mechanisms disrupting airway epithelial barrier during inflammation and reflect on whether these factors can be specifically targeted by repurposed existing drugs. Literature search was conducted using PubMed, drug database of US FDA and European Medicines Agency until and including September 2021. EXPERT OPINION IL-4 and IL-13 signaling are the major pathways disrupting the airway epithelial barrier during airway inflammation. However, blocking IL-4/IL-13 signaling may adversely affect protective immune responses and increase susceptibility of host to infections. An alternate approach to modulate airway epithelial barrier function involves targeting specific downstream component of IL-4/IL-13 signaling or different inflammatory mediators responsible for regulation of airway epithelial barrier. Airway epithelium-targeted therapy using inhibitors of HDAC, HSP90, MIF, mTOR, IL-17A and VEGF may be a potential strategy to prevent airway epithelial barrier dysfunction in airway inflammatory diseases.
Collapse
Affiliation(s)
- Ahsan Anjoom Sunil
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Tom Skaria
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
34
|
Cadmium and Cadmium/BDE (47 or 209) Exposure Affect Mitochondrial Function, DNA Damage/Repair Mechanisms and Barrier Integrity in Airway Epithelial Cells. ATMOSPHERE 2022. [DOI: 10.3390/atmos13020201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Heavy metals and Brominated diphenyl ether flame-retardants (BDEs) often coexist in the environment and are capable of inducing injury, cytotoxicity or genotoxicity in human epithelial cells of the lung. We studied the effects of single Cadmium chloride (CdCl2) or CdCl2/BDE (47 or 209) mixtures in airway epithelial cells, using A549 cell line cultured at submerged conditions and air–liquid interface (ALI) (an in vitro model described as physiologically relevant in vivo-like). We evaluated cell viability, oxidative stress, apoptosis, DNA damage/repair (Comet assay, γH2AX phosphorylation ser139), mitochondrial redox balance (NOX-4, Nrf2 and TFAM) and cell barrier integrity (TEER, ZO-1, Claudin-1, E-cadherin-1) in A549 cells exposed to CdCl2 (1 nM to 10 µM), or to CdCl2 (100 nM)/BDEs (47 or 209) (100 nM). CdCl2 (10 μM) reduced cell viability and increased apoptosis. CdCl2 (100 nM) significantly affected DNA-damage/repair (Olive Tail length production), γH2AX phosphorylation and oxidative stress (ROS/JC-1 production) in submerged cell cultures. CdCl2 (100 nM) decreased viability, TEER, ZO-1, Claudin-1 and E-cadherin-1 mRNA expression, and Nrf2 and TFAM while increased NOX-4, in ALI culture of cells. In both cell culture approaches, the cells stimulated with Cadmium/BDEs mixtures did not show a significant increase in the effects observed in the cells treated with CdCl2 alone. CdCl2 inhalation might exert cytotoxicity and genotoxicity, playing a pivotal role in the uncontrolled oxidative stress, damaging DNA and gene expression in airway epithelial cells. No additional or synergistic adverse effects of CdCl2/BDEs mixture were observed in comparison to CdCl2 alone in lung epithelium.
Collapse
|
35
|
Shah SA, Ishinaga H, Takeuchi K. Distinct Secretion of MUC5AC and MUC5B in Upper and Lower Chronic Airway Diseases. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.8060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The human airway is protected by a defensive mucus barrier. The most prominent components of mucus are the mucin glycoproteins MUC5AC and MUC5B. They are produced by goblet cells and submucosal gland cells in the upper and lower airways. Hyperplasia of these cells and hypersecretion of MUC5AC and MUC5B characterize chronic inflammatory diseases of the upper and lower airways. Recent studies have revealed that MUC5AC and MUC5B are expressed at specific sites in the respiratory tract through different molecular mechanisms and have distinct functions. Morphometric and histochemical studies have also examined the roles of goblet cells, submucosal gland cells, MUC5AC, and MUC5B in different chronic airway diseases individually. The individual study of goblet cells, submucosal gland cells, MUC5AC, and MUC5B in airway diseases would be helpful for precisely diagnosing chronic inflammatory diseases of the airway and establishing optimal treatments. This review focuses on the distinct secretion of MUC5AC and MUC5B and their producing cells in chronic inflammatory diseases of the upper and lower airway.
Collapse
|
36
|
Abstract
Viruses are essentially, obligate intracellular parasites. They require a host to replicate their genetic material, spread to other cells, and eventually to other hosts. For humans, most viral infections are not considered lethal, regardless if at the cellular level, the virus can obliterate individual cells. Constant genomic mutations, (which can alter the antigenic content of viruses such as influenza or coronaviruses), zoonosis or immunosuppression/immunocompromisation, is when viruses achieve higher host mortality. Frequent examples of the severe consequenses of viral infection can be seen in children and the elderly. In most instances, the immune system will take a multifaceted approach in defending the host against viruses. Depending on the virus, the individual, and the point of entry, the immune system will initiate a robust response which involves multiple components. In this chapter, we expand on the total immune system, breaking it down to the two principal types: Innate and Adaptive Immunity, their different roles in viral recognition and clearance. Finally, how different viruses activate and evade different arms of the immune system.
Collapse
|
37
|
Ruysseveldt E, Martens K, Steelant B. Airway Basal Cells, Protectors of Epithelial Walls in Health and Respiratory Diseases. FRONTIERS IN ALLERGY 2021; 2:787128. [PMID: 35387001 PMCID: PMC8974818 DOI: 10.3389/falgy.2021.787128] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
The airway epithelium provides a critical barrier to the outside environment. When its integrity is impaired, epithelial cells and residing immune cells collaborate to exclude pathogens and to heal tissue damage. Healing is achieved through tissue-specific stem cells: the airway basal cells. Positioned near the basal membrane, airway basal cells sense and respond to changes in tissue health by initiating a pro-inflammatory response and tissue repair via complex crosstalks with nearby fibroblasts and specialized immune cells. In addition, basal cells have the capacity to learn from previous encounters with the environment. Inflammation can indeed imprint a certain memory on basal cells by epigenetic changes so that sensitized tissues may respond differently to future assaults and the epithelium becomes better equipped to respond faster and more robustly to barrier defects. This memory can, however, be lost in diseased states. In this review, we discuss airway basal cells in respiratory diseases, the communication network between airway basal cells and tissue-resident and/or recruited immune cells, and how basal cell adaptation to environmental triggers occurs.
Collapse
Affiliation(s)
- Emma Ruysseveldt
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Katleen Martens
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Brecht Steelant
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Head and Neck Surgery, Department of Otorhinolaryngology, University of Crete School of Medicine, Heraklion, Greece
| |
Collapse
|
38
|
Mastalerz M, Dick E, Chakraborty AA, Hennen E, Schamberger AC, Schröppel A, Lindner M, Hatz R, Behr J, Hilgendorff A, Schmid O, Staab-Weijnitz CA. Validation of in vitro models for smoke exposure of primary human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2021; 322:L129-L148. [PMID: 34668416 DOI: 10.1152/ajplung.00091.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RATIONALE The bronchial epithelium is constantly challenged by inhalative insults including cigarette smoke (CS), a key risk factor for lung disease. In vitro exposure of bronchial epithelial cells using CS extract (CSE) is a widespread alternative to whole CS (wCS) exposure. However, CSE exposure protocols vary considerably between studies, precluding direct comparison of applied doses. Moreover, they are rarely validated in terms of physiological response in vivo and the relevance of the findings is often unclear. METHODS We tested six different exposure settings in primary human bronchial epithelial cells (phBECs), including five CSE protocols in comparison with wCS exposure. We quantified cell-delivered dose and directly compared all exposures using expression analysis of 10 well-established smoke-induced genes in bronchial epithelial cells. CSE exposure of phBECs was varied in terms of differentiation state, exposure route, duration of exposure, and dose. Gene expression was assessed by quantitative Real-Time PCR (qPCR) and Western Blot analysis. Cell type-specific expression of smoke-induced genes was analyzed by immunofluorescent analysis. RESULTS Three surprisingly dissimilar exposure types, namely chronic CSE treatment of differentiating phBECs, acute CSE treatment of submerged basal phBECs, and wCS exposure of differentiated phBECs performed best, resulting in significant upregulation of seven (chronic CSE) and six (acute wCS, acute submerged CSE exposure) out of 10 genes. Acute apical or basolateral exposure of differentiated phBECs with CSE was much less effective despite similar doses used. CONCLUSIONS Our findings provide guidance for the design of human in vitro CS exposure models in experimental and translational lung research.
Collapse
Affiliation(s)
- Michal Mastalerz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elisabeth Dick
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ashesh Anjankumar Chakraborty
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elisabeth Hennen
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andrea C Schamberger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andreas Schröppel
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | | | - Rudolf Hatz
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine, Viszeral-, Transplantations-, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Jürgen Behr
- Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität (LMU), Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Otmar Schmid
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Claudia A Staab-Weijnitz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
39
|
Burgoyne RA, Fisher AJ, Borthwick LA. The Role of Epithelial Damage in the Pulmonary Immune Response. Cells 2021; 10:cells10102763. [PMID: 34685744 PMCID: PMC8534416 DOI: 10.3390/cells10102763] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pulmonary epithelial cells are widely considered to be the first line of defence in the lung and are responsible for coordinating the innate immune response to injury and subsequent repair. Consequently, epithelial cells communicate with multiple cell types including immune cells and fibroblasts to promote acute inflammation and normal wound healing in response to damage. However, aberrant epithelial cell death and damage are hallmarks of pulmonary disease, with necrotic cell death and cellular senescence contributing to disease pathogenesis in numerous respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and coronavirus disease (COVID)-19. In this review, we summarise the literature that demonstrates that epithelial damage plays a pivotal role in the dysregulation of the immune response leading to tissue destruction and abnormal remodelling in several chronic diseases. Specifically, we highlight the role of epithelial-derived damage-associated molecular patterns (DAMPs) and senescence in shaping the immune response and assess their contribution to inflammatory and fibrotic signalling pathways in the lung.
Collapse
Affiliation(s)
- Rachel Ann Burgoyne
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Andrew John Fisher
- Regenerative Medicine, Stem Cells and Transplantation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Lee Anthony Borthwick
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Fibrofind, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Correspondence: ; Tel.: +44-191-208-3112
| |
Collapse
|
40
|
Montalbano AM, Chiappara G, Albano GD, Ferraro M, Di Sano C, Vitulo P, Pipitone L, Ricciardolo FLM, Anzalone G, Profita M. Expression/Activation of PAR-1 in Airway Epithelial Cells of COPD Patients: Ex Vivo/In Vitro Study. Int J Mol Sci 2021; 22:ijms221910703. [PMID: 34639044 PMCID: PMC8509732 DOI: 10.3390/ijms221910703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The role of PAR-1 expression and activation was described in epithelial cells from the central and distal airways of COPD patients using an ex vivo/in vitro model. PAR-1 immunoreactivity was studied in epithelial cells from surgical specimens of the central and distal airways of COPD patients and healthy control (HC). Furthermore, PAR-1 expression and activation were measured in both the human bronchial epithelial cell line (16HBE) and normal human bronchial epithelial cells (NHBEs) exposed to cigarette smoke extract (CSE) (10%) or thrombin. Finally, cell proliferation, apoptosis, and IL-8 release were detected in stimulated NHBEs. We identified higher levels of PAR-1 expression/activation in epithelial cells from the central airways of COPD patients than in HC. Active PAR-1 increased in epithelial cells from central and distal airways of COPD, with higher levels in COPD smokers (correlated with pack-years) than in COPD ex-smokers. 16HBE and NHBEs exposed to CSE or thrombin showed increased levels of active PAR-1 (localized in the cytoplasm) than baseline conditions, while NHBEs treated with thrombin or CSE showed increased levels of IL-8 proteins, with an additional effect when used in combination. Smoking habits generate the upregulation of PAR-1 expression/activation in airway epithelial cells, and promoting IL-8 release might affect the recruitment of infiltrating cells in the airways of COPD patients.
Collapse
Affiliation(s)
- Angela Marina Montalbano
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Giuseppina Chiappara
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Giusy Daniela Albano
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Maria Ferraro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Caterina Di Sano
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Patrizio Vitulo
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), 90127 Palermo, Italy; (P.V.); (L.P.)
| | - Loredana Pipitone
- Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (ISMETT), 90127 Palermo, Italy; (P.V.); (L.P.)
| | | | - Giulia Anzalone
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
| | - Mirella Profita
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90146 Palermo, Italy; (A.M.M.); (G.C.); (G.D.A.); (M.F.); (C.D.S.); (G.A.)
- Correspondence:
| |
Collapse
|
41
|
Viola H, Washington K, Selva C, Grunwell J, Tirouvanziam R, Takayama S. A High-Throughput Distal Lung Air-Blood Barrier Model Enabled By Density-Driven Underside Epithelium Seeding. Adv Healthc Mater 2021; 10:e2100879. [PMID: 34174173 DOI: 10.1002/adhm.202100879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Indexed: 12/18/2022]
Abstract
High-throughput tissue barrier models can yield critical insights on how barrier function responds to therapeutics, pathogens, and toxins. However, such models often emphasize multiplexing capability at the expense of physiologic relevance. Particularly, the distal lung's air-blood barrier is typically modeled with epithelial cell monoculture, neglecting the substantial contribution of endothelial cell feedback in the coordination of barrier function. An obstacle to establishing high-throughput coculture models relevant to the epithelium/endothelium interface is the requirement for underside cell seeding, which is difficult to miniaturize and automate. Therefore, this paper describes a scalable, low-cost seeding method that eliminates inversion by optimizing medium density to float cells so they attach under the membrane. This method generates a 96-well model of the distal lung epithelium-endothelium barrier with serum-free, glucocorticoid-free air-liquid differentiation. The polarized epithelial-endothelial coculture exhibits mature barrier function, appropriate intercellular junction staining, and epithelial-to-endothelial transmission of inflammatory stimuli such as polyinosine:polycytidylic acid (poly(I:C)). Further, exposure to influenza A virus PR8 and human beta-coronavirus OC43 initiates a dose-dependent inflammatory response that propagates from the epithelium to endothelium. While this model focuses on the air-blood barrier, the underside seeding method is generalizable to various coculture tissue models for scalable, physiologic screening.
Collapse
Affiliation(s)
- Hannah Viola
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology 311 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| | - Kendra Washington
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Cauviya Selva
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Jocelyn Grunwell
- Division of Critical Care Medicine Children's Healthcare of Atlanta at Egleston 1405 Clifton Road NE Atlanta GA 30322 USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics Emory University School of Medicine and Center for CF & Airways Disease Research 2015 Uppergate Dr NE, Rm 344 Atlanta GA 30322 USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| |
Collapse
|
42
|
Xu J, Meng Y, Jia M, Jiang J, Yang Y, Ou Y, Wu Y, Yan X, Huang M, Adcock IM, Yao X. Epithelial expression and role of secreted STC1 on asthma airway hyperresponsiveness through calcium channel modulation. Allergy 2021; 76:2475-2487. [PMID: 33378582 DOI: 10.1111/all.14727] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/16/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Asthma is characterized by airway hyperresponsiveness (AHR), inflammation, and airway remodeling. Airway hyperresponsiveness results from enhanced airway smooth muscle (ASM) contraction potentially under the control of an epithelium-derived relaxing factor (EpDRF). However, relatively rare is known about EpDRF. We aimed to elucidate the role of epithelium-derived stanniocalcin-1 (STC1) on AHR and ASM contraction. METHODS Stanniocalcin-1 levels in the serum of asthmatic patients and healthy volunteers and in bronchoalveolar lavage fluid (BALF) from ovalbumin (OVA)-challenged mice were measured by ELISA. The effects of exogenous STC1 on AHR and on inflammation were examined in mice. IL-13 modulation of STC1 mRNA and protein levels was studied in human bronchial epithelial cell lines (16HBE). The function of STC1 on Ca2+ influx and ASM contraction was examined ex vivo. RESULTS Serum STC1 was decreased in asthma (n = 93) compared with healthy volunteers (1071 ± 30.4 vs 1414 ± 75.1 pg/ml, p < 0.0001, n = 23) and correlated with asthma control (p = 0.0270), lung function (FEV1, p = 0.0130), and serum IL-13 levels (p = 0.0009). Treatment of ten asthmatic subjects with inhaled corticosteroids/long-acting beta2-agonists (ICS/LABA) for 1 year enhanced STC1 expression which correlated with improved asthma control (p = 0.022). STC1 was mainly expressed in bronchial epithelium and intranasal administration of recombinant human STC1 (rhSTC1) reduced AHR and inflammation in mice. IL-13 suppressed STC1 release from 16HBE, whereas rhSTC1 blocked store-operated Ca2+ entry (SOCE) by suppressing stromal interaction molecule 1 (STIM1) and further inhibited ASM cell contractility by suppressing Ca2+ -dependent myosin light chain (MLC) phosphorylation. CONCLUSION Our data indicate that STC1 deficiency in asthmatic airways promotes STIM1 hyperactivity, enhanced ASM contraction, and AHR. STC1 may be a candidate EpDRF.
Collapse
Affiliation(s)
- Jiayan Xu
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of Respiratory & Critical Care Medicine Northern Jiangsu People's Hospital Yangzhou China
| | - Yaqi Meng
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Man Jia
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Jie Jiang
- Department of Respiratory & Critical Care Medicine Huai'an First People's Hospital Huai'an China
| | - Yi Yang
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Yingwei Ou
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Yunhui Wu
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Xiaoyi Yan
- Department of Respiratory & Critical Care Medicine Nanjing Jiangning People's Hospital Nanjing China
| | - Mao Huang
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Ian M. Adcock
- Airway Disease Section Faculty of Medicine National Heart and Lung Institute Imperial College London London UK
| | - Xin Yao
- Department of Respiratory & Critical Care Medicine The First Affiliated Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
43
|
Zhang Y, Wang X, Zhang H, Tang H, Hu H, Wang S, Wong VKW, Li Y, Deng J. Autophagy Modulators From Chinese Herbal Medicines: Mechanisms and Therapeutic Potentials for Asthma. Front Pharmacol 2021; 12:710679. [PMID: 34366865 PMCID: PMC8342996 DOI: 10.3389/fphar.2021.710679] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/28/2021] [Indexed: 01/21/2023] Open
Abstract
Asthma has become a global health issue, suffering more than 300 million people in the world, which is a heterogeneous disease, usually characterized by chronic airway inflammation and airway hyperreactivity. Combination of inhaled corticosteroids (ICS) and long acting β-agonists (LABA) can relieve asthma symptoms and reduce the frequency of exacerbations, especially for patients with refractory asthma, but there are limited treatment options for people who do not gain control on combination ICS/LABA. The increase in ICS dose generally provides little additional benefit, and there is an increased risk of side effects. Therefore, therapeutic interventions integrating the use of different agents that focus on different targets are needed to overcome this set of diseases. Some findings suggest autophagy is closely correlated with the severity of asthma through eosinophilic inflammation, and its modulation may provide novel therapeutic approaches for severe allergic asthma. The chinese herbal medicine (CHM) have been demonstrated clinically as potent therapeutic interventions for asthma. Moreover some reports have found that the bioactive components isolated from CHM could modulate autophagy, and exhibit potent Anti-inflammatory activity. These findings have implied the potential for CHMs in asthma or allergic inflammation therapy via the modulation of autophagy. In this review, we discuss the basic pathomechanisms underpinning asthma, and the potential role of CHMs in treating asthma with modulating autophagy.
Collapse
Affiliation(s)
- Yun Zhang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xing Wang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - He Zhang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hongmei Tang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hang Hu
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Songping Wang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuying Li
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Deng
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
44
|
Pereira ABM, Oliveira JR, Souza ALJ, Andrade-Silva L, Silva MV, Silva PR, Silva-Vergara ML, Rogerio AP. Effects of cigarette smoke extract on bronchial epithelial cells stimulated with Cryptococcus neoformans. Med Microbiol Immunol 2021; 210:221-233. [PMID: 34228244 DOI: 10.1007/s00430-021-00715-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
In the airways, the adhesion of Cryptococcus neoformans with airway epithelial cells is crucial for the establishment of cryptococcosis. Tobacco smoke is considered a risk factor for cryptococcosis. Here, we evaluated the effects of cigarette smoke extract (CSE) on human bronchial epithelial cells (BEAS-2B) stimulated with C. neoformans. Multiplicities of infection (MOIs) of 1-100 of C. neoformans per cell led to increased IL-8 production and no cytotoxic effects when compared to those of controls. C. neoformans (MOI 100) also significantly increased the concentration of IL-6. In cells stimulated with CSE doses (1.0, 2.5 and 5.0%) from one or five cigarettes, increased IL-1β production was observed only in doses from one (1.0%) and five (2.5%) cigarettes when compared to that of controls. However, only 1.0% CSE failed to show cytotoxic effects. In addition, CSE significantly increased the concentration of IL-8. Cells stimulated with both CSE and C. neoformans demonstrated a reduction in IL-6/STAT3 signalling compared to that in cells stimulated by C. neoformans. In addition, a significant increase in IL-10 production was also observed. No alterations in NF-kB or ICAM-1 expression were observed among the groups. The combination of CSE and C. neoformans favoured the increase of fungal numbers and extracellular adhering of C. neoformans on BEAS-2B cells. In addition, the internalization of C. neoformans on BEAS-2B cells was reduced after CSE stimulation. In conclusion, the association of CSE and C. neoformans induced an anti-inflammatory effect in bronchial epithelial cells, which might favour the development of C. neoformans infection in the airways.
Collapse
Affiliation(s)
- Aline Beatriz Mahler Pereira
- Laboratory of Experimental Immunopharmacology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Street Vigário Carlos 162, Uberaba, MG, 38025-380, Brazil
| | - Jhony Robison Oliveira
- Laboratory of Experimental Immunopharmacology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Street Vigário Carlos 162, Uberaba, MG, 38025-380, Brazil
| | - Ana Leticia Julio Souza
- Laboratory of Experimental Immunopharmacology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Street Vigário Carlos 162, Uberaba, MG, 38025-380, Brazil
| | - Leonardo Andrade-Silva
- Laboratory of Mycology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Marcos Vinicius Silva
- Laboratory of Immunology, Institute Department of Clinical Medicine, of Health Sciences, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Paulo Roberto Silva
- Laboratory of Experimental Immunopharmacology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Street Vigário Carlos 162, Uberaba, MG, 38025-380, Brazil
| | - Mario Leon Silva-Vergara
- Laboratory of Mycology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Uberaba, MG, Brazil
| | - Alexandre Paula Rogerio
- Laboratory of Experimental Immunopharmacology, Department of Clinical Medicine, Institute of Health Sciences, Federal University of Triangulo Mineiro, Street Vigário Carlos 162, Uberaba, MG, 38025-380, Brazil.
| |
Collapse
|
45
|
Worrell JC, MacLeod MKL. Stromal-immune cell crosstalk fundamentally alters the lung microenvironment following tissue insult. Immunology 2021; 163:239-249. [PMID: 33556186 PMCID: PMC8014587 DOI: 10.1111/imm.13319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/22/2021] [Accepted: 02/03/2021] [Indexed: 12/21/2022] Open
Abstract
Communication between stromal and immune cells is essential to maintain tissue homeostasis, mount an effective immune response and promote tissue repair. This 'crosstalk' occurs in both the steady state and following a variety of insults, for example, in response to local injury, at sites of infection or cancer. What do we mean by crosstalk between cells? Reciprocal activation and/or regulation occurs between immune and stromal cells, by direct cell contact and indirect mechanisms, including the release of soluble cytokines. Moving beyond cell-to-cell contact, this review investigates the complexity of 'cross-space' cellular communication. We highlight different examples of cellular communication by a variety of lung stromal and immune cells following tissue insults. This review examines how the 'geography of the lung microenvironment' is altered in various disease states; more specifically, we investigate how this influences lung epithelial cells and fibroblasts via their communication with immune cells and each other.
Collapse
Affiliation(s)
- Julie C. Worrell
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Megan K. L. MacLeod
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| |
Collapse
|
46
|
Wang A, Zhang Q, Wang Y, Li X, Li K, Li Y, Wang J, Li L, Chen H. Inhibition of Gabrp reduces the differentiation of airway epithelial progenitor cells into goblet cells. Exp Ther Med 2021; 22:720. [PMID: 34007329 PMCID: PMC8120639 DOI: 10.3892/etm.2021.10152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/12/2021] [Indexed: 12/05/2022] Open
Abstract
Bronchial asthma is an intractable pulmonary disease that affects millions of individuals worldwide, with the overproduction of mucus contributing to high morbidity and mortality. Gamma-aminobutyric acid (GABA) is associated with goblet cell hyperplasia in the lungs of primate models and Club cells serve as airway epithelial progenitor cells that may differentiate into goblet and ciliated cells. In the present study, it was investigated whether the GABAA receptor pi (Gabrp) is essential for Club cell proliferation and differentiation in mice. Validation of microarray analysis results by reverse transcription-quantitative PCR (RT-qPCR) demonstrated that Gabrp is highly expressed in mouse Club cells. Predominant expression of Gabrp in mouse Club cells was further confirmed based on naphthalene-induced Club cell injury in mice, with organoid cultures indicating significant reductions in the organoid-forming ability of mouse Club cells in the presence of Gabrp antagonist bicuculline methiodide (BMI). Furthermore, the RT-qPCR results indicated that the mRNA levels of chloride channel accessory 3, pseudogene (Clca3p), mucin (Muc)5Ac and Muc5B were significantly decreased in BMI organoid cultures. These results suggested that blocking GABA signaling through Gabrp inhibits mouse Club cell proliferation, as well as differentiation into goblet cells. Therefore, targeting GABA/Gabrp signaling may represent a promising strategy for treating goblet cell hyperplasia in bronchial asthma.
Collapse
Affiliation(s)
- An Wang
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China
| | - Qiuyang Zhang
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
| | - Yongmei Wang
- Department of Pathology, Tianjin University Haihe Hospital, Tianjin 300350, P.R. China
| | - Xue Li
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
| | - Kuan Li
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
| | - Yu Li
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
| | - Jianhai Wang
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China
| | - Li Li
- Department of Respiratory Medicine, Tianjin University Haihe Hospital, Tianjin 300350, P.R. China
| | - Huaiyong Chen
- Department of Basic Medicine, Haihe Clinical College of Tianjin Medical University, Tianjin 300350, P.R. China.,Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese Medicine, Tianjin Institute of Respiratory Diseases, Tianjin 300350, P.R. China.,Tianjin Key Laboratory of Lung Regenerative Medicine, Tianjin 300350, P.R. China
| |
Collapse
|
47
|
Xiong R, Wu Y, Wu Q, Muskhelishvili L, Davis K, Tripathi P, Chen Y, Chen T, Bryant M, Rosenfeldt H, Healy SM, Cao X. Integration of transcriptome analysis with pathophysiological endpoints to evaluate cigarette smoke toxicity in an in vitro human airway tissue model. Arch Toxicol 2021; 95:1739-1761. [PMID: 33660061 PMCID: PMC8113308 DOI: 10.1007/s00204-021-03008-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/16/2021] [Indexed: 01/04/2023]
Abstract
Exposure to cigarette smoke (CS) is a known risk factor in the pathogenesis of smoking-caused diseases, such as chronic obstructive pulmonary diseases (COPD) and lung cancer. To assess the effects of CS on the function and phenotype of airway epithelial cells, we developed a novel repeated treatment protocol and comprehensively evaluated the progression of key molecular, functional, and structural abnormalities induced by CS in a human in vitro air-liquid-interface (ALI) airway tissue model. Cultures were exposed to CS (diluted with 0.5 L/min, 1.0 L/min, and 4.0 L/min clean air) generated from smoking five 3R4F University of Kentucky reference cigarettes under the International Organization for Standardization (ISO) machine smoking regimen, every other day for 4 weeks (3 days per week, 40 min/day). By integrating the transcriptomics-based approach with the in vitro pathophysiological measurements, we demonstrated CS-mediated effects on oxidative stress, pro-inflammatory cytokines and matrix metalloproteinases (MMPs), ciliary function, expression and secretion of mucins, and squamous cell differentiation that are highly consistent with abnormalities observed in airways of smokers. Enrichment analysis on the transcriptomic profiles of the ALI cultures revealed key molecular pathways, such as xenobiotic metabolism, oxidative stress, and inflammatory responses that were perturbed in response to CS exposure. These responses, in turn, may trigger aberrant tissue remodeling, eventually leading to the onset of respiratory diseases. Furthermore, changes of a panel of genes known to be disturbed in smokers with COPD were successfully reproduced in the ALI cultures exposed to CS. In summary, findings from this study suggest that such an integrative approach may be a useful tool for identifying genes and adverse cellular events caused by inhaled toxicants, like CS.
Collapse
Affiliation(s)
- Rui Xiong
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yue Wu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR, 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hans Rosenfeldt
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Sheila M Healy
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
48
|
Stanke F, Janciauskiene S, Tamm S, Wrenger S, Raddatz EL, Jonigk D, Braubach P. Effect of Alpha-1 Antitrypsin on CFTR Levels in Primary Human Airway Epithelial Cells Grown at the Air-Liquid-Interface. Molecules 2021; 26:molecules26092639. [PMID: 33946490 PMCID: PMC8125203 DOI: 10.3390/molecules26092639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) gene is influenced by the fundamental cellular processes like epithelial differentiation/polarization, regeneration and epithelial–mesenchymal transition. Defects in CFTR protein levels and/or function lead to decreased airway surface liquid layer facilitating microbial colonization and inflammation. The SERPINA1 gene, encoding alpha1-antitrypsin (AAT) protein, is one of the genes implicated in CF, however it remains unknown whether AAT has any influence on CFTR levels. In this study we assessed CFTR protein levels in primary human lung epithelial cells grown at the air-liquid-interface (ALI) alone or pre-incubated with AAT by Western blots and immunohistochemistry. Histological analysis of ALI inserts revealed CFTR- and AAT-positive cells but no AAT-CFTR co-localization. When 0.5 mg/mL of AAT was added to apical or basolateral compartments of pro-inflammatory activated ALI cultures, CFTR levels increased relative to activated ALIs. This finding suggests that AAT is CFTR-modulating protein, albeit its effects may depend on the concentration and the route of administration. Human lung epithelial ALI cultures provide a useful tool for studies in detail how AAT or other pharmaceuticals affect the levels and activity of CFTR.
Collapse
Affiliation(s)
- Frauke Stanke
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Correspondence: ; Tel.: +49-511-5326722
| | - Sabina Janciauskiene
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Stephanie Tamm
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
| | - Sabine Wrenger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Ellen Luise Raddatz
- Department of Pediatric Pneumology, Neonatology and Allergology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.T.); (E.L.R.)
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Peter Braubach
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Centre for Lung Research, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; (S.J.); (S.W.); (D.J.); (P.B.)
- Institute of Pathology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
49
|
Innes E, Yiu HHP, McLean P, Brown W, Boyles M. Simulated biological fluids - a systematic review of their biological relevance and use in relation to inhalation toxicology of particles and fibres. Crit Rev Toxicol 2021; 51:217-248. [PMID: 33905298 DOI: 10.1080/10408444.2021.1903386] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The use of simulated biological fluids (SBFs) is a promising in vitro technique to better understand the release mechanisms and possible in vivo behaviour of materials, including fibres, metal-containing particles and nanomaterials. Applications of SBFs in dissolution tests allow a measure of material biopersistence or, conversely, bioaccessibility that in turn can provide a useful inference of a materials biodistribution, its acute and long-term toxicity, as well as its pathogenicity. Given the wide range of SBFs reported in the literature, a review was conducted, with a focus on fluids used to replicate environments that may be encountered upon material inhalation, including extracellular and intracellular compartments. The review aims to identify when a fluid design can replicate realistic biological conditions, demonstrate operation validation, and/or provide robustness and reproducibility. The studies examined highlight simulated lung fluids (SLFs) that have been shown to suitably replicate physiological conditions, and identify specific components that play a pivotal role in dissolution mechanisms and biological activity; including organic molecules, redox-active species and chelating agents. Material dissolution was not always driven by pH, and likewise not only driven by SLF composition; specific materials and formulations correspond to specific dissolution mechanisms. It is recommended that SLF developments focus on biological predictivity and if not practical, on better biological mimicry, as such an approach ensures results are more likely to reflect in vivo behaviour regardless of the material under investigation.
Collapse
Affiliation(s)
- Emma Innes
- Institute of Occupational Medicine (IOM), Edinburgh, UK
| | - Humphrey H P Yiu
- Chemical Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Polly McLean
- Institute of Occupational Medicine (IOM), Edinburgh, UK
| | - William Brown
- Institute of Occupational Medicine (IOM), Edinburgh, UK
| | | |
Collapse
|
50
|
Zhou J, Zhou XD, Xu R, Du XZ, Li Q, Li B, Zhang GY, Chen LX, Perelman JM, Kolosov VP. The Degradation of Airway Epithelial Tight Junctions in Asthma Under High Airway Pressure Is Probably Mediated by Piezo-1. Front Physiol 2021; 12:637790. [PMID: 33868003 PMCID: PMC8047413 DOI: 10.3389/fphys.2021.637790] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/02/2022] Open
Abstract
Full functioning of the airway physical barrier depends on cellular integrity, which is coordinated by a series of tight junction (TJ) proteins. Due to airway spasm, edema, and mucus obstruction, positive end-expiratory alveolar pressure (also termed auto-PEEP) is a common pathophysiological phenomenon, especially in acute asthma attack. However, the influence of auto-PEEP on small airway epithelial TJs is currently unclear. We performed studies to investigate the effect of extra pressure on small airway epithelial TJs and its mechanism. The results first confirmed that a novel mechanosensitive receptor, piezo-1, was highly expressed in the airway epithelium of asthmatic mice. Extra pressure induced the degradation of occludin, ZO-1 and claudin-18 in primary human small airway epithelial cells (HSAECs), resulting in a decrease in transepithelial electrical resistance (TER) and an increase in cell layer permeability. Through in vitro investigations, we observed that exogenous pressure stimulation could elevate the intracellular calcium concentration ([Ca2+]i) in HSAECs. Downregulation of piezo-1 with siRNA and pretreatment with BAPTA-AM or ALLN reduced the degradation of TJs and attenuated the impairment of TJ function induced by exogenous pressure. These findings indicate the critical role of piezo-1/[Ca2+]i/calpain signaling in the regulation of small airway TJs under extra pressure stimulation.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xian-Zhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Bin Li
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fengjie, Chongqing, China
| | - Guo-Yue Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Xiu Chen
- Department of Respiratory Medicine, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Juliy M Perelman
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| | - Victor P Kolosov
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| |
Collapse
|