1
|
Chen Z, Shang Y, Duan W, Zhu L, Ji X, Gong S, Xiang X. Androgens have therapeutic potential in T2 asthma by mediating METTL3 in bronchial epithelial cells. Int Immunopharmacol 2024; 143:113322. [PMID: 39369464 DOI: 10.1016/j.intimp.2024.113322] [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: 06/26/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/08/2024]
Abstract
Studies have shown that androgens can alleviate the symptoms of T2 asthma and are inversely correlated with the severity of allergic asthma. METTL3, a crucial component of m6A modification, mitigates the development of T2 asthma by inhibiting Th2 cell differentiation. However, the impact of androgens, such as dihydrotestosterone (DHT), on the progression of T2 asthma through METTL3 has yet to be investigated. At the clinical level, patients with T2 asthma exhibited reduced levels of DHT and METTL3 mRNA, along with increased levels of 17β-estradiol (E2). DHT and METTL3 were found to be negatively associated with the severity of T2 asthma, while E2 was positively associated with it. Administration of DHT and E2 in induced T2 asthma mouse models showed that DHT improved lung function, reduced airway inflammation, and inhibited Th2 cell differentiation. Interestingly, DHT reversed the damage to METTL3, whereas E2 had the opposite effect. In vitro studies of mouse bronchial epithelial cells (BECs) confirmed that METTL3-dependent m6A modification inhibited the T2 inflammatory response, and DHT inhibited Th2 cell differentiation in T2 asthma by promoting METTL3 expression in BECs. In conclusion, our study suggests that DHT has therapeutic potential for T2 asthma by regulating METTL3 in BECs.
Collapse
Affiliation(s)
- Zhifeng Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yulin Shang
- Ophthalmology and Otorhinolaryngology, Zigui County Traditional Chinese Medicine Hospital, 30 Pinghu Avenue, Zigui, Hubei 443600, China
| | - Wentao Duan
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital, 61 West Jiefang Road, Changsha, Hunan 410005, China
| | - Liming Zhu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital, 61 West Jiefang Road, Changsha, Hunan 410005, China
| | - Xiaoying Ji
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Guiyang, Guizhou 550004, China.
| | - Subo Gong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China.
| | - Xudong Xiang
- Department of Emergency, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
2
|
Shiratsuchi G, Konishi S, Yano T, Yanagihashi Y, Nakayama S, Katsuno T, Kashihara H, Tanaka H, Tsukita K, Suzuki K, Herawati E, Watanabe H, Hirai T, Yagi T, Kondoh G, Gotoh S, Tamura A, Tsukita S. Dual-color live imaging unveils stepwise organization of multiple basal body arrays by cytoskeletons. EMBO Rep 2024; 25:1176-1207. [PMID: 38316902 PMCID: PMC10933483 DOI: 10.1038/s44319-024-00066-0] [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: 05/03/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 02/07/2024] Open
Abstract
For mucociliary clearance of pathogens, tracheal multiciliated epithelial cells (MCCs) organize coordinated beating of cilia, which originate from basal bodies (BBs) with basal feet (BFs) on one side. To clarify the self-organizing mechanism of coordinated intracellular BB-arrays composed of a well-ordered BB-alignment and unidirectional BB-orientation, determined by the direction of BB to BF, we generated double transgenic mice with GFP-centrin2-labeled BBs and mRuby3-Cep128-labeled BFs for long-term, high-resolution, dual-color live-cell imaging in primary-cultured tracheal MCCs. At early timepoints of MCC differentiation, BB-orientation and BB-local alignment antecedently coordinated in an apical microtubule-dependent manner. Later during MCC differentiation, fluctuations in BB-orientation were restricted, and locally aligned BB-arrays were further coordinated to align across the entire cell (BB-global alignment), mainly in an apical intermediate-sized filament-lattice-dependent manner. Thus, the high coordination of the BB-array was established for efficient mucociliary clearance as the primary defense against pathogen infection, identifying apical cytoskeletons as potential therapeutic targets.
Collapse
Affiliation(s)
- Gen Shiratsuchi
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Satoshi Konishi
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Tomoki Yano
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | | | - Shogo Nakayama
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- RIKEN Center for Biosystems Dynamics Research, Hyogo, Japan
| | - Tatsuya Katsuno
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroka Kashihara
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Hiroo Tanaka
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- School of Medicine, Teikyo University, Tokyo, Japan
| | - Kazuto Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koya Suzuki
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Elisa Herawati
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
- Faculty of Mathematics and Natural Sciences, Universitas Sebelas Maret, Surakarta, Central Java, Indonesia
| | - Hitomi Watanabe
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Yagi
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Gen Kondoh
- Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shimpei Gotoh
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Atsushi Tamura
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
- School of Medicine, Teikyo University, Tokyo, Japan.
| | - Sachiko Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan.
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
3
|
Janas PP, Chauché C, Shearer P, Perona-Wright G, McSorley HJ, Schwarze J. Cold dispase digestion of murine lungs improves recovery and culture of airway epithelial cells. PLoS One 2024; 19:e0297585. [PMID: 38271372 PMCID: PMC10810513 DOI: 10.1371/journal.pone.0297585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
Airway epithelial cells (AECs) play a key role in maintaining lung homeostasis, epithelium regeneration and the initiation of pulmonary immune responses. To isolate and study murine AECs investigators have classically used short and hot (1h 37°C) digestion protocols. Here, we present a workflow for efficient AECs isolation and culture, utilizing long and cold (20h 4°C) dispase II digestion of murine lungs. This protocol yields a greater number of viable AECs compared to an established 1h 37°C dispase II digestion. Using a combination of flow cytometry and immunofluorescent microscopy, we demonstrate that compared to the established method, the cold digestion allows for recovery of a 3-fold higher number of CD45-CD31-EpCAM+ cells from murine lungs. Their viability is increased compared to established protocols, they can be isolated in larger numbers by magnetic-activated cell sorting (MACS), and they result in greater numbers of distal airway stem cell (DASC) KRT5+p63+ colonies in vitro. Our findings demonstrate that temperature and duration of murine lung enzymatic digestion have a considerable impact on AEC yield, viability, and ability to form colonies in vitro. We believe this workflow will be helpful for studying lung AECs and their role in the biology of lung.
Collapse
Affiliation(s)
- Piotr Pawel Janas
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Caroline Chauché
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Patrick Shearer
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Georgia Perona-Wright
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Henry J. McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jürgen Schwarze
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom
| |
Collapse
|
4
|
Kitano M, Hayashi Y, Ohnishi H, Okuyama H, Yoshimatsu M, Mizuno K, Kuwata F, Tada T, Kishimoto Y, Morita S, Omori K. Changes in the Proportion of Each Cell Type After hiPSC-Derived Airway Epithelia Transplantation. Cell Transplant 2024; 33:9636897241228026. [PMID: 38372247 PMCID: PMC10878204 DOI: 10.1177/09636897241228026] [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: 11/04/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 02/20/2024] Open
Abstract
No radical treatment is available for the regeneration of dysfunction and defects in airway epithelia. Artificial tracheae made of polypropylene and collagen sponge were used in clinical studies to reconstitute tracheae after resection. For early epithelialization of the luminal surface of the artificial trachea, a model was established, that is, an artificial trachea covered with human-induced pluripotent stem cell-derived airway epithelial cells (hiPSC-AECs) was transplanted into a tracheal defect in an immunodeficient rat. Unlike the cell types of hiPSC-derived cells that are currently used in clinical studies, AECs maintain tissues by proliferation and differentiation of basal cells into various cell types that constitute AECs constantly. Therefore, post-transplantation, the proportion of each cell type, such as ciliated and goblet cells, may change; however, no studies have examined this possibility. In this study, using our hiPSC-AEC-transplanted rat model, we investigated changes in the proportion of each cell type in hiPSC-AECs pre-transplantation and post-transplantation. As a result, the proportion of each cell type changed post-transplantation. The proportion of ciliated, basal, and club cells increased, and the proportion of goblet cells decreased post-transplantation. In addition, the proportion of each cell type in engrafted hiPSC-AECs is more similar to the proportion of each cell type in normal proximal airway tissue than the proportion of each cell type pre-transplantation. The results of this study are useful for the development of therapeutic techniques using hiPSC-AEC transplantation.
Collapse
Affiliation(s)
- Masayuki Kitano
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuyuki Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideaki Okuyama
- School of Communication Sciences and Disorders, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Keisuke Mizuno
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Kuwata
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Tada
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Chen Z, Shang Y, Yuan Y, He Y, Wasti B, Duan W, Ouyang R, Jia J, Xiao B, Zhang D, Zhang X, Li J, Chen B, Liu Y, Zeng Q, Ji X, Ma L, Liu S, Xiang X. MBD2 mediates Th17 cell differentiation by regulating MINK1 in Th17-dominant asthma. Front Genet 2022; 13:959059. [PMID: 36303542 PMCID: PMC9592806 DOI: 10.3389/fgene.2022.959059] [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: 06/01/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: .Asthma is a highly heterogeneous disease, and T-helper cell type 17 (Th17) cells play a pathogenic role in the development of non-T2 severe asthma. Misshapen like kinase 1 (MINK1) is involved in the regulation of Th17 cell differentiation, but its effect on severe asthma remains unclear. Our previous studies showed that methyl-CpG binding domain protein 2 (MBD2) expression was significantly increased in patients with Th17 severe asthma and could regulate Th17 cell differentiation. The aim of this study was to investigate how MBD2 interacts with MINK1 to regulate Th17 cell differentiation in Th17-dominant asthma.Materials and methods: Female C57BL/6 mice and bronchial epithelial cells (BECs) were used to establish mouse and cell models of Th17-dominant asthma, respectively. Flow cytometry was used to detect Th17 cell differentiation, and the level of IL-17 was detected by enzyme-linked immunosorbent assay (ELISA). Western blot and quantitative real-time PCR (qRT-PCR) were used to detect MBD2 and MINK1 expression. To investigate the role of MBD2 and MINK1 in Th17 cell differentiation in Th17-dominant asthma, the MBD2 and MINK1 genes were silenced or overexpressed by small interfering RNA and plasmid transfection.Results: Mouse and BEC models of Th17-dominant asthma were established successfully. The main manifestations were increased neutrophils in BALF, airway hyperresponsiveness (AHR), activated Th17 cell differentiation, and high IL-17 levels. The expression of MBD2 in lung tissues and BECs from the Th17-dominant asthma group was significantly increased, while the corresponding expression of MINK1 was significantly impaired. Through overexpression or silencing of MBD2 and MINK1 genes, we have concluded that MBD2 and MINK1 regulate Th17 cell differentiation and IL-17 release. Interestingly, MBD2 was also found to negatively regulate the expression of MINK1.Conclusion: Our findings have revealed new roles for MBD2 and MINK1, and provide new insights into epigenetic regulation of Th17-dominant asthma, which is dominated by neutrophils and Th17 cells. This study could lead to new therapeutic targets for patients with Th17-dominant asthma.
Collapse
Affiliation(s)
- Zhifeng Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yulin Shang
- Ophthalmology and Otorhinolaryngology, Zigui County Traditional Chinese Medicine Hospital, Zigui, Hubei, China
| | - Yu Yuan
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi He
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Binaya Wasti
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wentao Duan
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruoyun Ouyang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingsi Jia
- Department of Emergency, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bing Xiao
- Department of Emergency, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dongshan Zhang
- Department of Emergency, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiufeng Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jianmin Li
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Bolin Chen
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People’s Hospital, Changsha, Hunan, China
| | - Yi Liu
- Department of Respiratory Medicine, Zhuzhou City Central Hospital, Zhuzhou, Hunan, China
| | - Qingping Zeng
- Department of Respiratory and Critical Care Medicine, Longshan County People’s Hospital, Longshan, Hunan, China
| | - Xiaoying Ji
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- *Correspondence: Libing Ma, ; Shaokun Liu, ; Xudong Xiang,
| | - Shaokun Liu
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Libing Ma, ; Shaokun Liu, ; Xudong Xiang,
| | - Xudong Xiang
- Department of Emergency, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Libing Ma, ; Shaokun Liu, ; Xudong Xiang,
| |
Collapse
|
6
|
Androgen Plays a Potential Novel Hormonal Therapeutic Role in Th17 Cells Predominant Neutrophilic Severe Asthma by Attenuating BECs Regulated Th17 Cells Differentiation via MBD2 Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3096528. [PMID: 36062195 PMCID: PMC9436621 DOI: 10.1155/2022/3096528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/02/2022] [Indexed: 12/05/2022]
Abstract
T helper 17 (Th17) cells subtype of non-T2 asthma is less responsive (resistant) to inhaled corticosteroids (ICS), so also called severe asthma. Methyl-CpG-binding domain protein 2 (MBD2) regulates the differentiation of the Th17 cells, showing the possibility of a therapeutic target in severe asthma. Androgen tends to show beneficial therapeutic effects and is a “hot research topic,” but its role in the differentiation and expression of Th17 cells via MBD2 is still unknown. The aim of this study was to evaluate how sex hormone interacts with MBD2 and affects the differentiation and expression of Th17 cells in severe asthma. Here, first, we measured the concentration of androgen, estrogen, and androgen estrogen ratio from subjects and correlated it with severe asthma status. Then, we established an animal model and bronchial epithelial cells (BECs) model of severe asthma to evaluate the role of MBD2 in the differentiation and expression of Th17 cells (IL-17), the therapeutic potential of sex hormones in severe asthma, and the effect of sex hormones in BECs regulated Th17 cells differentiation via MBD2 at the cellular level. Increased MBD2 expression and Th17 cells differentiation were noted in the animal and the BECs severe asthma models. Th17 cell differentiation and expression were MBD2 dependent. Androgen attenuated the differentiation of BECs regulated Th17 cells via MBD2 showing BECs as a therapeutic target of androgen, and these findings postulate the novel role of androgen in Th17 cells predominant neutrophilic severe asthma therapy through targeting MBD2.
Collapse
|
7
|
Hou Y, Ding Y, Du D, Yu T, Zhou W, Cui Y, Nie H. Airway Basal Cells Mediate Hypoxia-Induced EMT by Increasing Ribosome Biogenesis. Front Pharmacol 2021; 12:783946. [PMID: 34955855 PMCID: PMC8696177 DOI: 10.3389/fphar.2021.783946] [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: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 01/11/2023] Open
Abstract
Excessive secretion of airway mucus and fluid accumulation are the common features of many respiratory diseases, which, in turn, induce cell hypoxia in the airway epithelium, resulting in epithelial–mesenchymal transition (EMT) and ultimately fibrosis. However, the mechanisms of EMT induced by hypoxia in the airway are currently unclear. To mimic the status of edematous fluid retention in the airway, we cultured primary mouse tracheal epithelial cells (MTECs) in a liquid–liquid interface (LLI) mode after full differentiation in a classic air–liquid interface (ALI) culture system. The cell hypoxia was verified by the physical characteristics and lactate production in cultured medium as well as HIF expression in MTECs cultured by LLI mode. EMT was evidenced and mainly mediated by basal cells, supported by flow cytometry and immunofluorescence assay. The differently expressed genes of basal and other airway epithelial cells were found to be enriched in the ribosome by our analysis of an MTEC single-cell RNA sequencing data set and Myc, the global regulator of ribosome biogenesis was identified to be highly expressed in basal cells. We next separated basal cells from bulk MTECs by flow cytometry, and the real-time PCR results showed that ribosome biogenesis was significantly upregulated in basal cells, whereas the inhibition of ribosome biogenesis alleviated the phosphorylation of the mammalian target of rapamycin/AKT and abrogated hypoxia-induced EMT in MTECs. Collectively, these observations strongly suggest that basal cells in the airway epithelium may mediate the process of hypoxia-induced EMT, partly through enhancing ribosome biogenesis.
Collapse
Affiliation(s)
- Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Danni Du
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Wu Q, Fang L, Yang Y, Wang A, Chen X, Sun J, Wan J, Hong C, Tong J, Tao S, Tian H. Protection of melatonin against long-term radon exposure-caused lung injury. ENVIRONMENTAL TOXICOLOGY 2021; 36:472-483. [PMID: 33107683 DOI: 10.1002/tox.23052] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 09/04/2020] [Accepted: 10/18/2020] [Indexed: 06/11/2023]
Abstract
Radon is one of the major pathogenic factors worldwide. Recently, epidemiological studies have suggested that radon exposure plays an important role in lung injury, which could further cause cancer. However, the toxic effects and underlying mechanism on lung injury are still not clear. Here, we identified the detailed toxic effects of long-term radon exposure. Specifically, the manifestations were inflammatory response and cell apoptosis in dose- and time-dependent manners. In detail, it caused the mitochondrial dysfunction and oxidative stress as determined by the abnormal levels of mitochondrial DNA copy number, adenosine triphosphate, mitochondrial membrane potential, superoxide dismutase, and cycloxygenase-2. Furthermore, we found that melatonin treatment ameliorated mitochondrial dysfunction and attenuated the levels of oxidative stress caused by long-term radon exposure, which could further inhibit the lung tissue apoptosis as determined by the decreased levels of cleaved caspase 3. Our study would provide potential therapeutic application of melatonin on lung tissue injury caused by long-term radon exposure.
Collapse
Affiliation(s)
- Qianqian Wu
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Lijun Fang
- Shanghai Minhang District Center for Disease Prevention and Control, Shanghai, China
| | - Youjing Yang
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Aiqing Wang
- Medical College, Soochow University, Suzhou, China
| | - Xiaoyu Chen
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jiaojiao Sun
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jianmei Wan
- Medical College, Soochow University, Suzhou, China
| | | | - Jian Tong
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shasha Tao
- School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Hailin Tian
- School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| |
Collapse
|
9
|
Hou Y, Zhou Z, Liu H, Zhang H, Ding Y, Cui Y, Nie H. Mesenchymal Stem Cell-Conditioned Medium Rescues LPS-Impaired ENaC Activity in Mouse Trachea via WNK4 Pathway. Curr Pharm Des 2021; 26:3601-3607. [PMID: 32003683 DOI: 10.2174/1381612826666200131141732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Airway epithelium plays an essential role in maintaining the homeostasis and function of respiratory system as the first line of host defense. Of note, epithelial sodium channel (ENaC) is one of the victims of LPS-induced airway injury. Regarding the great promise held by mesenchymal stem cells (MSCs) for regenerative medicine in the field of airway injury and the limitations of cell-based MSCs therapy, we focused on the therapeutic effect of MSCs conditioned medium (MSCs-CM) on the ENaC activity in mouse tracheal epithelial cells. METHODS Ussing chamber apparatus was applied to record the short-circuit currents in primary cultured mouse tracheal epithelial cells, which reflects the ENaC activity. Expressions of α and γ ENaC were measured at the protein and mRNA levels by western blot and real-time PCR, respectively. The expression of with-no-lysinekinase- 4 (WNK4) and ERK1/2 were measured at protein levels, and the relationship between WNK4 and ERK1/2 was determined by WNK4 knockdown. RESULTS MSCs-CM restored the LPS-impaired ENaC activity, as well as enhanced the mRNA and protein expressions of ENaC in primary cultured mouse tracheal epithelial cells. Meanwhile, WNK4 and ERK1/2, both negative-regulators of ENaC, were suppressed accordingly after the administration of MSCs-CM in LPS-induced airway injury. After WNK4 gene was knocked down by siRNA, the level of ERK1/2 phosphorylation decreased. CONCLUSION In light of the key role of ENaC in fluid reabsorption and the beneficial effects of MSCs-CM in the injury of airway epithelium, our results suggest that MSCs-CM is effective in alleviating LPS-induced ENaC dysfunction through WNK4-ERK1/2 pathway, which will provide a potent direction for the therapy of airway injury.
Collapse
Affiliation(s)
- Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhiyu Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongfei Liu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
10
|
Chang J, Chen Z, Zhao R, Nie HG, Ji HL. Ion transport mechanisms for smoke inhalation-injured airway epithelial barrier. Cell Biol Toxicol 2020; 36:571-589. [PMID: 32588239 DOI: 10.1007/s10565-020-09545-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 06/17/2020] [Indexed: 10/24/2022]
Abstract
Smoke inhalation injury is the leading cause of death in firefighters and victims. Inhaled hot air and toxic smoke are the predominant hazards to the respiratory epithelium. We aimed to analyze the effects of thermal stress and smoke aldehyde on the permeability of the airway epithelial barrier. Transepithelial resistance (RTE) and short-circuit current (ISC) of mouse tracheal epithelial monolayers were digitized by an Ussing chamber setup. Zonula occludens-1 tight junctions were visualized under confocal microscopy. A cell viability test and fluorescein isothiocyanate-dextran assay were performed. Thermal stress (40 °C) decreased RTE in a two-phase manner. Meanwhile, thermal stress increased ISC followed by its decline. Na+ depletion, amiloride (an inhibitor for epithelial Na+ channels [ENaCs]), ouabain (a blocker for Na+/K+-ATPase), and CFTRinh-172 (a blocker of cystic fibrosis transmembrane regulator [CFTR]) altered the responses of RTE and ISC to thermal stress. Steady-state 40 °C increased activity of ENaCs, Na+/K+-ATPase, and CFTR. Acrolein, one of the main oxidative unsaturated aldehydes in fire smoke, eliminated RTE and ISC. Na+ depletion, amiloride, ouabain, and CFTRinh-172 suppressed acrolein-sensitive ISC, but showed activating effects on acrolein-sensitive RTE. Thermal stress or acrolein disrupted zonula occludens-1 tight junctions, increased fluorescein isothiocyanate-dextran permeability but did not cause cell death or detachment. The synergistic effects of thermal stress and acrolein exacerbated the damage to monolayers. In conclusion, the paracellular pathway mediated by the tight junctions and the transcellular pathway mediated by active and passive ion transport pathways contribute to impairment of the airway epithelial barrier caused by thermal stress and acrolein. Graphical abstract Thermal stress and acrolein are two essential determinants for smoke inhalation injury, impairing airway epithelial barrier. Transcellular ion transport pathways via the ENaC, CFTR, and Na/K-ATPase are interrupted by both thermal stress and acrolein, one of the most potent smoke toxins. Heat and acrolein damage the integrity of the airway epithelium through suppressing and relocating the tight junctions.
Collapse
Affiliation(s)
- Jianjun Chang
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA.,Institute of Health Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Zaixing Chen
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA.,School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning, China
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA
| | - Hong-Guang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, 11937 US Hwy 271, Tyler, TX, 75708, USA. .,Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA.
| |
Collapse
|
11
|
Hou Y, Cui Y, Zhou Z, Liu H, Zhang H, Ding Y, Nie H, Ji HL. Upregulation of the WNK4 Signaling Pathway Inhibits Epithelial Sodium Channels of Mouse Tracheal Epithelial Cells After Influenza A Infection. Front Pharmacol 2019; 10:12. [PMID: 30723408 PMCID: PMC6349759 DOI: 10.3389/fphar.2019.00012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/07/2019] [Indexed: 12/28/2022] Open
Abstract
Influenza virus has a significant impact on the respiratory system. The mechanism of how influenza virus impairs the fluid transport in airway is not fully understood. We examined its effects on epithelial sodium channels (ENaC), which are very important for water and salt transport in the respiratory system. We focused on the impacts of influenza virus on ENaC activity in mouse tracheal epithelial cells (MTECs) and applied Ussing chamber apparatus for recording the short-circuit currents in primary cultured MTECs. Expressions of α and γ-ENaC were measured at the protein and mRNA levels by western blot and quantitative real-time polymerase chain reaction, respectively. Roles of the with-no-lysine-kinase-4 (WNK4) pathway were considered in participating influenza virus-involved ENaC regulation by using siRNA to knockdown WNK4 and the physical properties of airway surface liquid (ASL) were detected by confocal microscopy. Our results showed that influenza virus reduced ENaC activity, and the expressions of α and γ-ENaC were decreased at the protein and mRNA levels, respectively. WNK4 expression increased time-dependently at the protein level after influenza virus infection, while knockdown of WNK4 rescued the impact of influenza virus on ENaC and ASL height increased obviously after MTECs were treated with influenza virus. Taken together, these results suggest that influenza virus causes the changes of biophysical profile in the airway by altering the ENaC activity at least partly via facilitating the expression of WNK4.
Collapse
Affiliation(s)
- Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhou
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Hongfei Liu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Honglei Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States.,Texas Lung Injury Institute, The University of Texas Health Northeast, Tyler, TX, United States
| |
Collapse
|
12
|
Abstract
Epithelial abnormalities underpin the development of the middle ear disease, otitis media (OM). Until now, a well-characterized in vitro model of the middle ear (ME) epithelium that replicates the complex cellular composition of the middle ear has not been available. This chapter describes the development of a novel in vitro model of mouse middle ear epithelial cells (mMECs), cultured at the air-liquid interface (ALI). This system enables recapitulation of the characteristics of the native murine ME epithelium. We demonstrate that mMECs undergo differentiation into the varied cell populations seen within the native middle ear. Overall, our mMEC culture system can help better understand the cell biology of the middle ear and improve our understanding of the pathophysiology of OM. The model also has the potential to serve as a platform for validation of treatments designed to reverse aspects of epithelial remodeling underpinning OM development.
Collapse
|
13
|
Gkatzis K, Taghizadeh S, Huh D, Stainier DYR, Bellusci S. Use of three-dimensional organoids and lung-on-a-chip methods to study lung development, regeneration and disease. Eur Respir J 2018; 52:13993003.00876-2018. [PMID: 30262579 DOI: 10.1183/13993003.00876-2018] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 09/16/2018] [Indexed: 11/05/2022]
Abstract
Differences in lung anatomy between mice and humans, as well as frequently disappointing results when using animal models for drug discovery, emphasise the unmet need for in vitro models that can complement animal studies and improve our understanding of human lung physiology, regeneration and disease. Recent papers have highlighted the use of three-dimensional organoids and organs-on-a-chip to mimic tissue morphogenesis and function in vitro Here, we focus on the respiratory system and provide an overview of these in vitro models, which can be derived from primary lung cells and pluripotent stem cells, as well as healthy or diseased lungs. We emphasise their potential application in studies of respiratory development, regeneration and disease modelling.
Collapse
Affiliation(s)
- Konstantinos Gkatzis
- Dept of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sara Taghizadeh
- Dept of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Dongeun Huh
- Dept of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Didier Y R Stainier
- Dept of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Saverio Bellusci
- Dept of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Excellence Cluster Cardio-Pulmonary System, Justus-Liebig University Giessen, Giessen, Germany and German Center for Lung Research (DZL)
| |
Collapse
|
14
|
TRPA1 channels: expression in non-neuronal murine lung tissues and dispensability for hyperoxia-induced alveolar epithelial hyperplasia. Pflugers Arch 2018; 470:1231-1241. [PMID: 29754249 DOI: 10.1007/s00424-018-2148-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/22/2018] [Accepted: 04/27/2018] [Indexed: 01/31/2023]
Abstract
Transient receptor potential A1 (TRPA1) channels were originally characterized in neuronal tissues but also identified in lung epithelium by staining with fluorescently coupled TRPA1 antibodies. Its exact function in non-neuronal tissues, however, is elusive. TRPA1 is activated in vitro by hypoxia and hyperoxia and is therefore a promising TRP candidate for sensing hyperoxia in pulmonary epithelial cells and for inducing alveolar epithelial hyperplasia. Here, we isolated tracheal, bronchial, and alveolar epithelial cells and show low but detectable TRPA1 mRNA levels in all these cells as well as TRPA1 protein by Western blotting in alveolar type II (AT II) cells. We quantified changes in intracellular Ca2+ ([Ca2+]i) levels induced by application of hyperoxic solutions in primary tracheal epithelial, bronchial epithelial, and AT II cells isolated from wild-type (WT) and TRPA1-deficient (TRPA1-/-) mouse lungs. In all cell types, we detected hyperoxia-induced rises in [Ca2+]i levels, which were not significantly different in TRPA1-deficient cells compared to WT cells. We also tested TRPA1 function in a mouse model for hyperoxia-induced alveolar epithelial hyperplasia. A characteristic significant increase in thickening of alveolar tissues was detected in mouse lungs after exposure to hyperoxia, but not in normoxic WT and TRPA1-/- controls. Quantification of changes in lung morphology in hyperoxic WT and TRPA1-/- mice, however, again revealed no significant changes. Therefore, TRPA1 expression does neither appear to be a key player for hyperoxia-induced changes in [Ca2+]i levels in primary lung epithelial cells, nor being essential for the development of hyperoxia-induced alveolar epithelial hyperplasia.
Collapse
|
15
|
Song DJ, Miller M, Beppu A, Rosenthal P, Das S, Karta M, Vuong C, Mehta AK, Croft M, Broide DH. Rhinovirus Infection of ORMDL3 Transgenic Mice Is Associated with Reduced Rhinovirus Viral Load and Airway Inflammation. THE JOURNAL OF IMMUNOLOGY 2017; 199:2215-2224. [PMID: 28827284 DOI: 10.4049/jimmunol.1601412] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 07/27/2017] [Indexed: 01/29/2023]
Abstract
Orosomucoid like 3 (ORMDL3), a gene localized to chromosome 17q21, has been linked in epidemiologic studies to childhood asthma and rhinovirus (RV) infections. As the single nucleotide polymorphisms linking ORMDL3 to asthma are associated with increased expression of ORMDL3, we have used hORMDL3zp3-Cre mice (which have universal increased expression of human ORMDL3) to determine whether infection of these transgenic mice with RV influences levels of airway inflammation or RV viral load. RV infection of hORMDL3zp3-Cre mice resulted in reduced RV viral load assessed by quantitative real-time PCR (lung and airway epithelium), as well as reduced airway inflammation (total bronchoalveolar lavage cells, neutrophils, macrophages, and lymphocytes) compared with RV-infected wild-type mice. Levels of the antiviral pathways including IFNs (IFN-α, IFN-β, IFN-λ) and RNAse L were significantly increased in the lungs of RV-infected hORMDL3zp3-Cre mice. Levels of the antiviral mouse oligoadenylate synthetase (mOas)1g pathway and RNAse L were upregulated in the lungs of unchallenged hORMDL3zp3-Cre mice. In addition, levels of mOas2, but not mOas1 (mOas1a, mOas1b, mOas1g), or mOas3 pathways were significantly more upregulated by IFNs (IFN-α, IFN-β, IFN-λ) in epithelial cells from hORMDL3zp3-Cre mice compared with RV-infected wild-type mouse epithelial cells. RNAse L-deficient mice infected with RV had increased RV viral load. Overall, these studies suggest that increased levels of ORMDL3 contribute to antiviral defense to RV infection in mice through pathways that may include IFNs (IFN-α, IFN-β, IFN-λ), OAS, and RNAse L.
Collapse
Affiliation(s)
- Dae Jin Song
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093.,Department of Pediatrics, Korea University College of Medicine, Seoul 03080, Korea; and
| | - Marina Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Andrew Beppu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter Rosenthal
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sudipta Das
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Maya Karta
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Christine Vuong
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Amit Kumar Mehta
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute, La Jolla, CA 92037
| | - David H Broide
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
| |
Collapse
|
16
|
Hahn A, Faulhaber J, Srisawang L, Stortz A, Salomon JJ, Mall MA, Frings S, Möhrlen F. Cellular distribution and function of ion channels involved in transport processes in rat tracheal epithelium. Physiol Rep 2017; 5:e13290. [PMID: 28642338 PMCID: PMC5492199 DOI: 10.14814/phy2.13290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 01/17/2023] Open
Abstract
Transport of water and electrolytes in airway epithelia involves chloride-selective ion channels, which are controlled either by cytosolic Ca2+ or by cAMP The contributions of the two pathways to chloride transport differ among vertebrate species. Because rats are becoming more important as animal model for cystic fibrosis, we have examined how Ca2+- dependent and cAMP- dependent Cl- secretion is organized in the rat tracheal epithelium. We examined the expression of the Ca2+-gated Cl- channel anoctamin 1 (ANO1), the cystic fibrosis transmembrane conductance regulator (CFTR) Cl- channel, the epithelial Na+ channel ENaC, and the water channel aquaporin 5 (AQP5) in rat tracheal epithelium. The contribution of ANO1 channels to nucleotide-stimulated Cl- secretion was determined using the channel blocker Ani9 in short-circuit current recordings obtained from primary cultures of rat tracheal epithelial cells in Ussing chambers. We found that ANO1, CFTR and AQP5 proteins were expressed in nonciliated cells of the tracheal epithelium, whereas ENaC was expressed in ciliated cells. Among nonciliated cells, ANO1 occurred together with CFTR and Muc5b and, in addition, in a different cell type without CFTR and Muc5b. Bioelectrical studies with the ANO1-blocker Ani9 indicated that ANO1 mediated the secretory response to the nucleotide uridine-5'-triphosphate. Our data demonstrate that, in rat tracheal epithelium, Cl- secretion and Na+ absorption are routed through different cell types, and that ANO1 channels form the molecular basis of Ca2+-dependent Cl- secretion in this tissue. These characteristic features of Cl--dependent secretion reveal similarities and distinct differences to secretory processes in human airways.
Collapse
Affiliation(s)
- Anne Hahn
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johannes Faulhaber
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Lalita Srisawang
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Andreas Stortz
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Johanna J Salomon
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC) German Center for Lung Research (DZL) University of Heidelberg, Heidelberg, Germany
| | - Stephan Frings
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| | - Frank Möhrlen
- Department of Animal Molecular Physiology, Centre of Organismal Studies University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
17
|
Ng Kuet Leong N, Brombacher F, Dalpke AH, Weitnauer M. Crosstalk between glucocorticoids and IL-4 modulates Ym1 expression in alternatively activated myeloid cells. Immunobiology 2017; 222:759-767. [PMID: 28209270 DOI: 10.1016/j.imbio.2017.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/31/2022]
Abstract
Airway epithelial cells induce a tolerogenic microenvironment by modulating immune cells in the lung. We recently showed that the supernatant of airway epithelial cells induces two marker genes of alternative activation, Ym1 and Ms4a8a, in respiratory myeloid cells. This induction was partially mediated by glucocorticoids, secreted by airway epithelial cells. In this study, we further investigated Ym1 and Ms4a8a regulation in alternatively activated myeloid cells in the presence of the TH2 cytokines IL-4 and IL-13. We show that Ym1 expression is boosted upon co-stimulation with airway epithelial cell supernatant and IL-4/IL-13, whereas Ms4a8a expression is down-regulated. This suggests that a crosstalk between IL-4/IL-13 and glucocorticoid signaling exists. Blocking protein synthesis indicated that dexamethasone-induced de novo protein synthesis is required for the interaction between glucocorticoid and IL-4 signaling regarding Ym1 regulation. Using reporter gene constructs, we demonstrate that the important regulatory region within the Ym1 promoter is found between -602bp and -969bp upstream of the start of translation. Bioinformatic analysis identified several glucocorticoid response elements (GREs) in this region. Further analysis identified overlapping but functionally active glucocorticoid receptor and STAT-6 binding sites, supporting the cooperative effect of glucocorticoids and IL-4 in the regulation of Ym1. These findings further prove the plasticity and complexity of alternatively activated myeloid cells and the importance of the local microenvironment. We believe that this regulation is of special importance in the pulmonary system, since both factors, glucocorticoids and IL-4/13, play a role in airway diseases such as asthma.
Collapse
Affiliation(s)
- Nathalie Ng Kuet Leong
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB) & Institute of Infectious Disease and Molecular Medicine (IDM), Division of Immunology, University of Cape Town & South African Medical Research Council (SAMRC), South Africa
| | - Alexander H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL).
| | - Michael Weitnauer
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
18
|
Zimmer J, Weitnauer M, Boutin S, Küblbeck G, Thiele S, Walker P, Lasitschka F, Lunding L, Orinska Z, Vock C, Arnold B, Wegmann M, Dalpke A. Nuclear Localization of Suppressor of Cytokine Signaling-1 Regulates Local Immunity in the Lung. Front Immunol 2016; 7:514. [PMID: 27917175 PMCID: PMC5114302 DOI: 10.3389/fimmu.2016.00514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/04/2016] [Indexed: 01/14/2023] Open
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a negative feedback inhibitor of cytoplasmic Janus kinase and signal transducer and activator of transcription (STAT) signaling. SOCS1 also contains a nuclear localization sequence (NLS), yet, the in vivo importance of nuclear translocation is unknown. We generated transgenic mice containing mutated Socs1ΔNLS that fails to translocate in the cell nucleus (MGLtg mice). Whereas mice fully deficient for SOCS1 die within the first 3 weeks due to excessive interferon signaling and multiorgan inflammation, mice expressing only non-nuclear Socs1ΔNLS (Socs1-/-MGLtg mice) were rescued from early lethality. Canonical interferon gamma signaling was still functional in Socs1-/-MGLtg mice as shown by unaltered tyrosine phosphorylation of STAT1 and whole genome expression analysis. However, a subset of NFκB inducible genes was dysregulated. Socs1-/-MGLtg mice spontaneously developed low-grade inflammation in the lung and had elevated Th2-type cytokines. Upon ovalbumin sensitization and challenge, airway eosinophilia was increased in Socs1-/-MGLtg mice. Decreased transepithelial electrical resistance in trachea epithelial cells from Socs1-/-MGLtg mice suggests disrupted epithelial cell barrier. The results indicate that nuclear SOCS1 is a regulator of local immunity in the lung and unravel a so far unrecognized function for SOCS1 in the cell nucleus.
Collapse
Affiliation(s)
- Jana Zimmer
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg , Heidelberg , Germany
| | - Michael Weitnauer
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg , Heidelberg , Germany
| | - Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany; German Center for Lung Research (DZL), Germany
| | | | - Sabrina Thiele
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg , Heidelberg , Germany
| | - Patrick Walker
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg , Heidelberg , Germany
| | - Felix Lasitschka
- Institute of Pathology, University Hospital Heidelberg , Heidelberg , Germany
| | - Lars Lunding
- German Center for Lung Research (DZL), Germany; Division of Asthma Mouse Model, Research Center Borstel, Borstel, Germany; Airway Research Center North, Borstel, Germany
| | - Zane Orinska
- German Center for Lung Research (DZL), Germany; Airway Research Center North, Borstel, Germany; Division of Experimental Pneumology, Prority Area Asthma & Allergy, Research Center Borstel, Borstel, Germany
| | - Christina Vock
- German Center for Lung Research (DZL), Germany; Airway Research Center North, Borstel, Germany; Division of Experimental Pneumology, Prority Area Asthma & Allergy, Research Center Borstel, Borstel, Germany
| | - Bernd Arnold
- German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Michael Wegmann
- German Center for Lung Research (DZL), Germany; Division of Asthma Mouse Model, Research Center Borstel, Borstel, Germany; Airway Research Center North, Borstel, Germany
| | - Alexander Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, University Hospital Heidelberg, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany; German Center for Lung Research (DZL), Germany
| |
Collapse
|
19
|
Mulay A, Akram KM, Williams D, Armes H, Russell C, Hood D, Armstrong S, Stewart JP, Brown SDM, Bingle L, Bingle CD. An in vitro model of murine middle ear epithelium. Dis Model Mech 2016; 9:1405-1417. [PMID: 27660200 PMCID: PMC5117233 DOI: 10.1242/dmm.026658] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/15/2016] [Indexed: 01/01/2023] Open
Abstract
Otitis media (OM), or middle ear inflammation, is the most common paediatric disease and leads to significant morbidity. Although understanding of underlying disease mechanisms is hampered by complex pathophysiology it is clear that epithelial abnormalities underpin the disease. There is currently a lack of a well-characterised in vitro model of the middle ear (ME) epithelium that replicates the complex cellular composition of the middle ear. Here, we report the development of a novel in vitro model of mouse middle ear epithelial cells (mMECs) at an air–liquid interface (ALI) that recapitulates the characteristics of the native murine ME epithelium. We demonstrate that mMECs undergo differentiation into the varied cell populations seen within the native middle ear. Proteomic analysis confirmed that the cultures secrete a multitude of innate defence proteins from their apical surface. We showed that the mMECs supported the growth of the otopathogen, nontypeable Haemophilus influenzae (NTHi), suggesting that the model can be successfully utilised to study host–pathogen interactions in the middle ear. Overall, our mMEC culture system can help to better understand the cell biology of the middle ear and improve our understanding of the pathophysiology of OM. The model also has the potential to serve as a platform for validation of treatments designed to reverse aspects of epithelial remodelling that underpin OM development. Summary: Development and systematic characterisation of an in vitro otopathogenic infection model of the murine middle ear epithelium as a tool to better understand the complex pathophysiology of Otitis media.
Collapse
Affiliation(s)
- Apoorva Mulay
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK
| | - Khondoker M Akram
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK
| | | | - Hannah Armes
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK.,Oral and Maxillofacial Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - Catherine Russell
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK
| | - Derek Hood
- MRC Mammalian Genetics Unit, Harwell OX11 0RD, UK
| | - Stuart Armstrong
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK
| | - James P Stewart
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L3 5RF, UK
| | | | - Lynne Bingle
- Oral and Maxillofacial Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK
| |
Collapse
|
20
|
Arsenic compromises conducting airway epithelial barrier properties in primary mouse and immortalized human cell cultures. PLoS One 2013; 8:e82970. [PMID: 24349408 PMCID: PMC3857810 DOI: 10.1371/journal.pone.0082970] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 11/08/2013] [Indexed: 12/31/2022] Open
Abstract
Arsenic is a lung toxicant that can lead to respiratory illness through inhalation and ingestion, although the most common exposure is through contaminated drinking water. Lung effects reported from arsenic exposure include lung cancer and obstructive lung disease, as well as reductions in lung function and immune response. As part of their role in innate immune function, airway epithelial cells provide a barrier that protects underlying tissue from inhaled particulates, pathogens, and toxicants frequently found in inspired air. We evaluated the effects of a five-day exposure to environmentally relevant levels of arsenic {<4μM [~300 μg/L (ppb)] as NaAsO2} on airway epithelial barrier function and structure. In a primary mouse tracheal epithelial (MTE) cell model we found that both micromolar (3.9 μM) and submicromolar (0.8 μM) arsenic concentrations reduced transepithelial resistance, a measure of barrier function. Immunofluorescent staining of arsenic-treated MTE cells showed altered patterns of localization of the transmembrane tight junction proteins claudin (Cl) Cl-1, Cl-4, Cl-7 and occludin at cell-cell contacts when compared with untreated controls. To better quantify arsenic-induced changes in tight junction transmembrane proteins we conducted arsenic exposure experiments with an immortalized human bronchial epithelial cell line (16HBE14o-). We found that arsenic exposure significantly increased the protein expression of Cl-4 and occludin as well as the mRNA levels of Cl-4 and Cl-7 in these cells. Additionally, arsenic exposure resulted in altered phosphorylation of occludin. In summary, exposure to environmentally relevant levels of arsenic can alter both the function and structure of airway epithelial barrier constituents. These changes likely contribute to the observed arsenic-induced loss in basic innate immune defense and increased infection in the airway.
Collapse
|
21
|
Repair of naphthalene-induced acute tracheal injury by basal cells depends on β-catenin. J Thorac Cardiovasc Surg 2013; 148:322-32. [PMID: 24280717 DOI: 10.1016/j.jtcvs.2013.10.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/25/2013] [Accepted: 10/11/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Little is known about the role of Wnt/β-catenin in postnatal airway homeostasis and basal cell function. This study aimed to investigate the role of Wnt signaling in the self-renewal of basal cells and the involvement of β-catenin in tracheal repair after naphthalene-induced injury. METHODS Mice were treated with naphthalene and injected with 4-hydroxytamoxifen. Injury and repair of the tracheal epithelium after naphthalene-mediated secretory cell depletion was assessed by a immunohistochemical study. The involvement of Wnt and β-catenin signaling in basal cell proliferation was investigated during in vitro expansion. RESULTS Immunohistochemical analysis of tracheal epithelium in wild-type mice showed a reduction in the number of Clara cell secretory protein (CCSP+) and forkhead box transcription factor (Fox-J1+) cells on days 2 to 5 after naphthalene-induced injury; this cell population was regenerated by day 10. After flush labeling, bromodeoxyuridine-positive (BrdU+) cells and Ki67+ cells were observed in tracheal epithelium on days 2 to 5 but not on days 10 and 21. Confocal microscopy visualizing K5+ and BrdU+ cells showed that Wnt3a promotes proliferation of K5+ cells. Immunohistochemical analysis of K5+ and CCSP+ in tracheal epithelial cells from wild-type littermate and K5-Cre-mediated β-catenin knock-out mice showed that on day 3, the number of CCSP+ cells was decreased in all mice. On day 10, CCSP+ cells were present in wild-type littermate mice but absent in conditional knock-out mice. CONCLUSIONS Basal cells serve as stem cells in the tracheal epithelium, regenerating and maintaining tracheal epithelial cells in a mouse model of tracheal injury. β-Catenin is required for proliferation and self-renewal of tracheal epithelial cells.
Collapse
|
22
|
Ovalbumin enhances YKL-40, IL-5, GM-CSF, and eotaxin expression simultaneously in primarily cultured mouse tracheal epithelial cells. In Vitro Cell Dev Biol Anim 2013; 50:243-50. [PMID: 24142153 DOI: 10.1007/s11626-013-9698-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
Epithelial inflammation and eosinophil infiltration are crucial for the pathogenesis of asthma. Many inflammatory mediators, such as YKL-40, interleukin -5 (IL-5), granulocyte-macrophage colony-stimulating factor (GM-CSF), and eotaxin, are important for the development of allergic airway inflammation. This study is aimed at investigating the impact of treatment with ovalbumin (OVA) on the levels of those inflammatory mediators in primarily cultured mouse tracheal epithelial cells. Mouse tracheal epithelial cells were isolated and identified by immunofluorescent staining; the isolated mouse tracheal epithelial cells expressed cytokeratins. Treatment with OVA for 24 or 48 h significantly increased the relative levels of YKL-40, IL-5, GM-CSF, and eotaxin mRNA transcripts and YKL-40, IL-5, GM-CSF, and eotaxin proteins secreted in the supernatants of cultured cells, as compared with that in the untreated control cells (P < 0.01, P < 0.05, respectively). The levels of YKL-40 expression were correlated positively with the levels of IL-5, GM-CSF, and eotaxin expression in the OVA-treated cells. These data indicated that treatment with OVA simultaneously enhanced YKL-40, IL-5, GM-CSF, and eotaxin expression in the cultured mouse tracheal epithelial cells in vitro. These inflammatory mediators may synergistically contribute to the pathogenesis of allergic inflammation, and this study may help to understand the role of YKL-40 in the pathogenesis of asthma.
Collapse
|
23
|
Abstract
Ion channels perform a variety of cellular functions in lung epithelia. Oxidant- and antioxidant-mediated mechanisms (that is, redox regulation) of ion channels are areas of intense research. Significant progress has been made in our understanding of redox regulation of ion channels since the last Experimental Biology report in 2003. Advancements include: 1) identification of nonphagocytic NADPH oxidases as sources of regulated reactive species (RS) production in epithelia, 2) an understanding that excessive treatment with antioxidants can result in greater oxidative stress, and 3) characterization of novel RS signaling pathways that converge upon ion channel regulation. These advancements, as discussed at the 2013 Experimental Biology Meeting in Boston, MA, impact our understanding of oxidative stress in the lung, and, in particular, illustrate that the redox state has profound effects on ion channel and cellular function.
Collapse
|
24
|
Flynn AN, Hoffman J, Tillu DV, Sherwood CL, Zhang Z, Patek R, Asiedu MNK, Vagner J, Price TJ, Boitano S. Development of highly potent protease-activated receptor 2 agonists via synthetic lipid tethering. FASEB J 2013; 27:1498-510. [PMID: 23292071 DOI: 10.1096/fj.12-217323] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Protease-activated receptor-2 (PAR₂) is a G-protein coupled receptor (GPCR) associated with a variety of pathologies. However, the therapeutic potential of PAR₂ is limited by a lack of potent and specific ligands. Following proteolytic cleavage, PAR₂ is activated through a tethered ligand. Hence, we reasoned that lipidation of peptidomimetic ligands could promote membrane targeting and thus significantly improve potency and constructed a series of synthetic tethered ligands (STLs). STLs contained a peptidomimetic PAR₂ agonist (2-aminothiazol-4-yl-LIGRL-NH₂) bound to a palmitoyl group (Pam) via polyethylene glycol (PEG) linkers. In a high-throughput physiological assay, these STL agonists displayed EC₅₀ values as low as 1.47 nM, representing a ∼200 fold improvement over the untethered parent ligand. Similarly, these STL agonists were potent activators of signaling pathways associated with PAR₂: EC₅₀ for Ca(2+) response as low as 3.95 nM; EC₅₀ for MAPK response as low as 9.49 nM. Moreover, STLs demonstrated significant improvement in potency in vivo, evoking mechanical allodynia with an EC₅₀ of 14.4 pmol. STLs failed to elicit responses in PAR2(-/-) cells at agonist concentrations of >300-fold their EC₅₀ values. Our results demonstrate that the STL approach is a powerful tool for increasing ligand potency at PAR₂ and represent opportunities for drug development at other protease activated receptors and across GPCRs.
Collapse
Affiliation(s)
- Andrea N Flynn
- Department of Physiology, Arizona Health Sciences Center, Tucson, AZ 85724-5030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sherwood CL, Lantz RC, Boitano S. Chronic arsenic exposure in nanomolar concentrations compromises wound response and intercellular signaling in airway epithelial cells. Toxicol Sci 2012. [PMID: 23204110 DOI: 10.1093/toxsci/kfs331] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Paracrine ATP signaling in the lung epithelium participates in a variety of innate immune functions, including mucociliary clearance, bactericide production, and as an initiating signal in wound repair. We evaluated the effects of chronic low-dose arsenic relevant to U.S. drinking water standards (i.e., 10 ppb [130nM]) on airway epithelial cells. Immortalized human bronchial epithelial cells (16HBE14o-) were exposed to 0, 130, or 330nM arsenic (as Na-arsenite) for 4-5 weeks and examined for wound repair efficiency and ATP-mediated Ca(2+) signaling. We found that chronic arsenic exposure at these low doses slows wound repair and reduces ATP-mediated Ca(2+) signaling. We further show that arsenic compromises ATP-mediated Ca(2+) signaling by altering both Ca(2+) release from intracellular stores (via metabotropic P2Y receptors) and Ca(2+) influx mechanisms (via ionotropic P2X receptors). To better model the effects of arsenic on ATP-mediated Ca(2+) signaling under conditions of natural exposure, we cultured tracheal epithelial cells obtained from mice exposed to control or 50 ppb Na-arsenite supplemented drinking water for 4 weeks. Tracheal epithelial cells from arsenic-exposed mice displayed reduced ATP-mediated Ca(2+) signaling dynamics similar to our in vitro chronic exposure. Our findings demonstrate that chronic arsenic exposure at levels that are commonly found in drinking water (i.e., 10-50 ppb) alters cellular mechanisms critical to airway innate immunity.
Collapse
Affiliation(s)
- Cara L Sherwood
- Arizona Respiratory Center, University of Arizona, Tucson, AZ, USA
| | | | | |
Collapse
|
26
|
Thevenot PT, Saravia J, Jin N, Giaimo JD, Chustz RE, Mahne S, Kelley MA, Hebert VY, Dellinger B, Dugas TR, Demayo FJ, Cormier SA. Radical-containing ultrafine particulate matter initiates epithelial-to-mesenchymal transitions in airway epithelial cells. Am J Respir Cell Mol Biol 2012; 48:188-97. [PMID: 23087054 DOI: 10.1165/rcmb.2012-0052oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Environmentally persistent free radicals (EPFRs) in combustion-generated particulate matter (PM) are capable of inducing pulmonary pathologies and contributing to the development of environmental asthma. In vivo exposure of infant rats to EPFRs demonstrates their ability to induce airway hyperresponsiveness to methacholine, a hallmark of asthma. However, the mechanisms by which combustion-derived EPFRs elicit in vivo responses remain elusive. In this study, we used a chemically defined EPFR consisting of approximately 0.2 μm amorphrous silica containing 3% cupric oxide with the organic pollutant 1,2-dichlorobenzene (DCB-230). DCB-230 possesses similar radical content to urban-collected EPFRs but offers several advantages, including lack of contaminants and chemical uniformity. DCB-230 was readily taken up by BEAS-2B and at high doses (200 μg/cm(2)) caused substantial necrosis. At low doses (20 μg/cm(2)), DCB-230 particles caused lysosomal membrane permeabilization, oxidative stress, and lipid peroxidation within 24 hours of exposure. During this period, BEAS-2B underwent epithelial-to-mesenchymal transition (EMT), including loss of epithelial cell morphology, decreased E-cadherin expression, and increased α-smooth muscle actin (α-SMA) and collagen I production. Similar results were observed in neonatal air-liquid interface culture (i.e., disruption of epithelial integrity and EMT). Acute exposure of infant mice to DCB-230 resulted in EMT, as confirmed by lineage tracing studies and evidenced by coexpression of epithelial E-cadherin and mesenchymal α-SMA proteins in airway cells and increased SNAI1 expression in the lungs. EMT in neonatal mouse lungs after EPFR exposure may provide an explanation for epidemiological evidence supporting PM exposure and increased risk of asthma.
Collapse
Affiliation(s)
- Paul T Thevenot
- Louisiana State University Health Sciences Center, Department of Pharmacology & Experimental Therapeutics, New Orleans, LA 70112, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Airway epithelial cell biology has been greatly advanced by studies of genetically defined and modified mice; however it is often difficult to isolate, manipulate, and assay epithelial cell-specific responses in vivo. In vitro proliferation and differentiation of mouse airway epithelial cells are made possible by a high-fidelity system for primary culture of mouse tracheal epithelial cells described in this chapter. Using this method, epithelial cells purified from mouse tracheas proliferate in growth factor-enriched medium. Subsequent culture in defined medium and the use of the air-liquid interface condition result in the development of well-differentiated epithelia composed of ciliated and non-ciliated cells with characteristics of native airways. Methods are also provided for manipulation of differentiation and analysis of differentiation and gene expression. These approaches allow the assessment of global responses and those of specific cell subpopulations within the airway epithelium.
Collapse
|
28
|
Taniguchi K, Yamamoto S, Aoki S, Toda S, Izuhara K, Hamasaki Y. Epigen is induced during the interleukin-13-stimulated cell proliferation in murine primary airway epithelial cells. Exp Lung Res 2011; 37:461-70. [PMID: 21867383 DOI: 10.3109/01902148.2011.596894] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Airway remodeling in bronchial asthma is characterized by epithelial detachment and proliferation, subepithelial fibrosis, increased smooth muscle mass, and goblet cell hyperplasia. These features are mediated by T-helper type 2 (Th2) cytokines including interleukin (IL)-13. However, the direct effects of IL-13 on the functions of airway epithelial cells are not fully understood. Murine primary airway epithelial (MPAE) cells were cultured in an air-liquid interface (ALI) culture system. AG1478, a specific inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase, was used to examine whether EGFR was involved in the IL-13-stimulated proliferation of MPAE cells. The expressions of EGFR ligands were investigated by reverse transcriptase-polymerase chain reaction, Western blotting, and immunohistochemical analyses. The cell counting in cross-sections and [(3)H]thymidine incorporation assays revealed a significant increase in the number of MPAE cells cultured with IL-13 compared with a phosphate-buffered saline (PBS) control. AG1478 abolished the IL-13-stimulated proliferation of MPAE cells. The expression of epigen, one of the EGFR ligands, was enhanced in MPAE cells cultured with IL-13. The findings suggest that EGFR is involved in the IL-13-stimulated proliferation of MPAE cells, and that epigen is important for the proliferation process in airway remodeling.
Collapse
|
29
|
Tate MD, Schilter HC, Brooks AG, Reading PC. Responses of mouse airway epithelial cells and alveolar macrophages to virulent and avirulent strains of influenza A virus. Viral Immunol 2011; 24:77-88. [PMID: 21449718 DOI: 10.1089/vim.2010.0118] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Influenza A virus (IAV) infection is associated with outcomes ranging from subclinical infection to severe pneumonia. In this study, we compared IAV strains BJx109 (H3N2), HKx31 (H3N2), and PR8 (H1N1), for their ability to elicit innate immune responses from mouse airway cells in vitro and their virulence in mice. The viruses differed markedly in their ability to induce disease in mice (PR8 > HKx31 > BJx109). In particular, PR8 infection was associated with high levels of virus replication and pulmonary inflammation. We next compared the ability of each virus strain to infect and induce inflammatory mediators from mouse airway cells. First, major differences were observed in the ability of viruses to infect and induce chemokines and cytokines from mouse alveolar macrophages (BJx109 > HKx31 > PR8), but not from airway epithelial cells (AEC) in vitro. Second, C-type lectins of the innate immune system in mouse lung fluids blocked the ability of BJx109, but not PR8, to infect mouse macrophages and AEC. The failure of the virulent PR8 virus to elicit responses from airway macrophages, combined with resistance to antiviral proteins in mouse airway fluids, likely contribute to virulence in mice. These findings provide insight into the mechanisms underlying disease severity in the mouse model of influenza infection.
Collapse
Affiliation(s)
- Michelle D Tate
- Department of Microbiology and Immunology, University of Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
30
|
Zhong Z, Wan Y, Han J, Shi S, Zhang Z, Sun X. Improvement of adenoviral vector-mediated gene transfer to airway epithelia by folate-modified anionic liposomes. Int J Nanomedicine 2011; 6:1083-93. [PMID: 21698075 PMCID: PMC3118681 DOI: 10.2147/ijn.s19745] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Indexed: 11/23/2022] Open
Abstract
Despite remarkable progress in the development of both viral and nonviral gene delivery vectors for airway disease treatment, poor gene transfer efficiency to the airway epithelium is a major obstacle in clinical application. To take advantage of the unique features of viral and nonviral vectors, we have developed complexes of adenovirus vector and anionic liposomes (AL-Ad5) by the calcium-induced phase change method. In the current study, based on the fact that there are overexpressed folate receptors on the surface of airway epithelia, we further modified the AL-Ad5 complexes with folate (F-AL-Ad5) to improve the transduction ability of Ad5 in airway epithelia. The transduction efficiencies of the obtained F-AL-Ad5 and AL-Ad5 complexes were assessed in primary-cultured airway epithelia in vitro. Our results indicated that compared with naked adenovirus vector, both AL-Ad5 and F-AL-Ad5 could significantly enhance the gene transduction efficiency of adenovirus vector in primary-cultured airway epithelial cells. Moreover, the enhancement mediated by F-AL-Ad5 was more dramatic than that by AL-Ad5. These results suggested that F-AL-Ad5 may be a useful strategy to deliver therapeutic genes to the airway epithelia and is promising in clinical application.
Collapse
Affiliation(s)
- Zhirong Zhong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | |
Collapse
|
31
|
Lam HC, Choi AMK, Ryter SW. Isolation of mouse respiratory epithelial cells and exposure to experimental cigarette smoke at air liquid interface. J Vis Exp 2011:2513. [PMID: 21372793 DOI: 10.3791/2513] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pulmonary epithelial cells can be isolated from the respiratory tract of mice and cultured at air-liquid interface (ALI) as a model of differentiated respiratory epithelium. A protocol is described for isolating and exposing these cells to mainstream cigarette smoke (CS), in order to study epithelial cell responses to CS exposure. The protocol consists of three parts: the isolation of airway epithelial cells from mouse trachea, the culturing of these cells at air-liquid interface (ALI) as fully differentiated epithelial cells, and the delivery of calibrated mainstream CS to these cells in culture. The ALI culture system allows the culture of respiratory epithelia under conditions that more closely resemble their physiological setting than ordinary liquid culture systems. The study of molecular and lung cellular responses to CS exposure is a critical component of understanding the impact of environmental air pollution on human health. Research findings in this area may ultimately contribute towards understanding the etiology of chronic obstructive pulmonary disease (COPD), and other tobacco-related diseases, which represent major global health problems.
Collapse
Affiliation(s)
- Hilaire C Lam
- Department of Medicine, Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, USA
| | | | | |
Collapse
|
32
|
Bharat A, Kuo E, Saini D, Steward N, Hachem R, Trulock EP, Patterson GA, Meyers BF, Mohanakumar T. Respiratory virus-induced dysregulation of T-regulatory cells leads to chronic rejection. Ann Thorac Surg 2010; 90:1637-44; discussion 1644. [PMID: 20971279 DOI: 10.1016/j.athoracsur.2010.06.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 06/01/2010] [Accepted: 06/07/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND Lower respiratory viral infections predispose to bronchiolitis obliterans syndrome (BOS). In addition, there is emerging evidence to support the role of autoimmunity in the pathogenesis of BOS. Because CD4(+)CD25(+)Foxp3(+) regulatory T-cells (Treg) control autoimmunity, we tested the hypothesis that respiratory virus-induced Treg dysfunction leads to BOS. METHODS Treg frequency was monitored using flow cytometry. Apoptosis, cytokines, and antibodies were analyzed using annexin V assay, LUMINEX, and enzyme-linked immunosorbent assay, respectively. Murine studies were performed using the orthotopic tracheal transplant model. RESULTS (A) Human studies: Treg troughs (decrease >50% of baseline) were found in 13 (43.3%) of 30 lung transplant recipients. Treg isolated during troughs revealed increased apoptosis (37.8%). Patients with Treg troughs had increased prevalence of antibodies to self-antigens collagen type I (23.1% vs 5.8% pretrough), collagen V (7.7% vs 0%), and k-alpha tubulin (30.7% vs 11.7%, p < 0.01) at 6 months post-trough. Increased number of Treg troughs correlated with more rapid onset of BOS. (B) Murine studies: Infection of tracheal transplant recipients with murine parainfleunza sendai virus led to increased Treg apoptosis (50.5%) in the draining lymph nodes. Vaccination against sendai virus prior to transplant abrogated apoptosis of Treg. In vitro, sendai virus-infected, but not naive, tracheal epithelial cells demonstrated upregulation of FasL (>3.5-fold) and induction of co-cultured Treg apoptosis (5.6-fold increase). CONCLUSIONS Respiratory viral infections cause Treg apoptosis which leads to the development of de novo autoimmunity that may play a role in the pathogenesis of BOS.
Collapse
Affiliation(s)
- Ankit Bharat
- Department of Surgery/Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Qiao J, Li A, Jin X. TSLP from RSV-stimulated rat airway epithelial cells activates myeloid dendritic cells. Immunol Cell Biol 2010; 89:231-8. [PMID: 20603637 DOI: 10.1038/icb.2010.85] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The respiratory syncytial virus (RSV) is a primary cause of lower respiratory tract infections in children, the elderly and in people who are immune suppressed, and is also the cause for the development of asthma primarily in infants. However, the immunological mechanisms by which RSV enhances allergic sensitization and asthma remain unclear. The aim of this study was to examine the influence of RSV-infected airway epithelial cells on the activation and functions of rat myeloid dendritic cells (mDCs).We found that the exposure of primary rat airway epithelial cells (PRAECs) to RSV induced a rapid (6 h), high (12 h) and persistent (18 h) increase in thymic stromal lymphopoietin (TSLP) mRNA compared with untreated PRAECs. TSLP protein expression was also enhanced by RSV infection. Functional maturation of mDCs was induced by RSV-treated PRAECs, as shown by their enhanced levels of OX40L and thymus- and activation-regulated chemokine (TARC) mRNAs, which increased the expressions of major histocompatibility complex II (MHCII) and CD86 costimulatory molecules and promoted enhanced T-cell proliferation in mixed lymphocyte reactions. These activities were inhibited in cocultures with RSV-infected RTECs (rat tracheal epithelial cells, an immortalized cell strain) that had been pretreated with TSLP-targeted small interfering RNA. These results suggest that RSV can induce epithelial cells to produce TSLP, which in turn promotes the maturation of mDCs that might support Th2 cell polarization.
Collapse
Affiliation(s)
- Jianou Qiao
- Respiratory Department, Shanghai Jiao Tong University Affiliated Ninth People's Hospital, Shanghai, China
| | | | | |
Collapse
|
34
|
Wu CA, Peluso JJ, Zhu L, Lingenheld EG, Walker ST, Puddington L. Bronchial epithelial cells produce IL-5: implications for local immune responses in the airways. Cell Immunol 2010; 264:32-41. [PMID: 20494340 DOI: 10.1016/j.cellimm.2010.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 04/19/2010] [Accepted: 04/26/2010] [Indexed: 01/09/2023]
Abstract
IL-5 is a pleiotropic cytokine that promotes eosinophil differentiation and survival. While naïve bronchial epithelial cells (BEC) produce low levels of IL-5, the role of BEC-derived IL-5 in allergic airway inflammation is unknown. We now show that BEC, isolated from mice with OVA-induced allergic airway disease (AAD), produced elevated levels of IL-5 mRNA and protein as compared to BEC from naïve mice. To determine the contribution of BEC-derived IL-5 to effector responses in the airways, IL-5 deficient bone marrow chimeric mice were generated in which IL-5 expression was restricted to stromal (e.g. BEC) or hematopoietic cells. When subjected to AAD, IL-5 produced by BECs contributed to mucous metaplasia, airway eosinophilia, and OVA-specific IgA levels. Thus, IL-5 production by BEC can impact the microenvironment of the lung, modifying pathologic and protective immune responses in the airways.
Collapse
Affiliation(s)
- Carol A Wu
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030-1319, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Duan H, Jiang Y, Zhang H, Wu Y. MiR-320 and miR-494 affect cell cycles of primary murine bronchial epithelial cells exposed to benzo[a]pyrene. Toxicol In Vitro 2010; 24:928-35. [DOI: 10.1016/j.tiv.2009.11.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 11/02/2009] [Accepted: 11/09/2009] [Indexed: 10/20/2022]
|
36
|
Gavilanes X, Huaux F, Meyer M, Lebecque P, Marbaix E, Lison D, Scholte B, Wallemacq P, Leal T. Azithromycin fails to reduce increased expression of neutrophil-related cytokines in primary-cultured epithelial cells from cystic fibrosis mice. J Cyst Fibros 2009; 8:203-10. [DOI: 10.1016/j.jcf.2009.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 02/11/2009] [Accepted: 03/03/2009] [Indexed: 02/08/2023]
|
37
|
Adherence and invasion of avian pathogenic Escherichia coli to avian tracheal epithelial cells. World J Microbiol Biotechnol 2009. [DOI: 10.1007/s11274-009-9978-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Brockman-Schneider RA, Amineva SP, Bulat MV, Gern JE. Serial culture of murine primary airway epithelial cells and ex vivo replication of human rhinoviruses. J Immunol Methods 2008; 339:264-9. [PMID: 18824171 DOI: 10.1016/j.jim.2008.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 08/22/2008] [Accepted: 09/02/2008] [Indexed: 10/21/2022]
Abstract
Human rhinoviruses (HRV) are the primary etiological agents in cold infections, and represent a serious risk to individuals with chronic respiratory disease such as asthma. In order to develop treatment options for HRV infections, murine models are a crucial component in the study of infection mechanisms due to the wide array of reagents and techniques available to study murine immunology. We present here a cell culture system for studying isolated murine epithelial cell responses to HRV. Monolayers of primary mouse airway epithelial cells were maintained in a serial culture system, and the identity and purity of the cell population was confirmed via immunostaining (positive for cytokeratin, negative for vimentin). Infection of these cells with a minor group rhinovirus (HRV-1A) was evidenced by increases in viral RNA, de novo synthesis of viral proteins, and production of infectious virus. This model will be useful in experiments to define mechanisms of viral replication and host/virus interactions within airway epithelial cells.
Collapse
|
39
|
Isolation and culture of primary equine tracheal epithelial cells. In Vitro Cell Dev Biol Anim 2008; 44:179-84. [PMID: 18594938 DOI: 10.1007/s11626-008-9099-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Accepted: 03/27/2008] [Indexed: 10/21/2022]
Abstract
Culture of airway epithelial cells is a useful model to investigate physiology of airway epithelia and airway disease mechanisms. In vitro models of airway epithelial cells are established for various species. However, earlier published method for isolation and culture of equine tracheal epithelial cells requires significant improvements. In this report, the development of a procedure for efficient isolation, characterization, culture, and passage of primary equine tracheal epithelial cells are described. Epithelial cells were isolated from adult equine trachea by exposing and stripping the mucosal epithelium from the adjacent connective tissue and smooth muscle. The tissue was minced and dissociated enzymatically using 0.25% trypsin-ethylenediaminetetraacetic acid (EDTA) solution for 2 h at 37 degrees C. Cells were collected by sieving and centrifugation, and contaminating fibroblasts were removed by differential adhesion. This procedure resulted in a typical yield of 1 x 10(7) cytokeratin-positive epithelial cells per gram tracheal lining tissue. Viability was 95% by trypan blue exclusion and isolates contained approximately 94% cytokeratin-positive cells of epithelial origin. Cells seeded at a density of 6.9 x 10(4) cells/cm2 in serum-free airway epithelial cell growth medium formed monolayers near confluency within a week. Confluent cells were dissociated using dispase II and first passages (P1) and second passages (P2) were successfully established in serum-free medium. Collagen coating of tissue culture flask was not required for cell adhesion, and cultures could be maintained at the level of P2 over 30 d. In the present study, we could establish a high-yield protocol for isolation and culture of equine tracheal epithelial cells that can serve for in vitro/ex vivo studies on the (patho-)physiology of equine airway disease as well as pharmacological and toxicological targets relevant to airway diseases.
Collapse
|
40
|
Liu Y, Wang Y, Jiang Y, Zhu N, Liang H, Xu L, Feng X, Yang H, Ma T. Mild processing defect of porcine DeltaF508-CFTR suggests that DeltaF508 pigs may not develop cystic fibrosis disease. Biochem Biophys Res Commun 2008; 373:113-8. [PMID: 18555011 DOI: 10.1016/j.bbrc.2008.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Accepted: 06/02/2008] [Indexed: 11/26/2022]
Abstract
Recent efforts have made significant progress in generating transgenic pigs with the DeltaF508-CFTR mutation to model the lung and pancreatic disease of human cystic fibrosis. However, species differences in the processing and function of human, pig and mouse DeltaF508-CFTR reported recently raise concerns about the phenotypic consequence of the gene-targeted pig model. The purpose of the present study was to characterize the DeltaF508 mutant of porcine CFTR to evaluate the severity of its processing defect. Biochemical and immunofluorescence analysis in transfected COS7 and FRT cells indicated that pig DeltaF508-CFTR efficiently targets to the plasma membrane and is present mainly as the mature glycosylated protein. Functional characterization in stably transfected FRT cells by fluorometric and electrophysiological assays supported efficient plasma membrane targeting of pig DeltaF508-CFTR. The mild cellular processing defect of pig DeltaF508-CFTR suggests that its gene-targeted pig model may not develop the lung and pancreatic phenotypes seen in CF patients.
Collapse
Affiliation(s)
- Yanli Liu
- Membrane Channel Research Laboratory, Northeast Normal University, 5268 Renmin Street, Changchun, Jilin 130024, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Mayer A, Bartz H, Fey F, Schmidt L, Dalpke A. Airway epithelial cells modify immune responses by inducing an anti-inflammatory microenvironment. Eur J Immunol 2008; 38:1689-99. [DOI: 10.1002/eji.200737936] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
42
|
Zaffuto KM, Estevez CN, Afonso CL. Primary chicken tracheal cell culture system for the study of infection with avian respiratory viruses. Avian Pathol 2008; 37:25-31. [PMID: 18202946 DOI: 10.1080/03079450701774850] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A major route of infection of avian influenza virus (AIV) and Newcastle disease virus (NDV) in chickens is through cells of the respiratory epithelium. Here we describe the development of a method for culture of tracheal epithelial cells from chicken embryos as well as their use in studies of infection with avian respiratory viruses such as low-pathogenicity AIV and lentogenic NDV. Positive immunostaining for cytokeratin, the presence of cilia and microvilli, and microarray analysis of transcribed RNA demonstrated that the isolated cells were epithelial in nature. Infection of the epithelial cell cultures with AIV and NDV was demonstrated using immunofluorescence or green fluorescence protein fluorescence microscopy, respectively. Growth curves of AIV and NDV in tracheal epithelial cells revealed that tracheal epithelial cells can fully support AIV and NDV growth and reinfection. This system, which mimics that of the natural infection, will be useful to study the mechanisms of early viral infection and cellular host transcriptional responses.
Collapse
Affiliation(s)
- K M Zaffuto
- Southeast Poultry Research Laboratory, USDA-ARS, Athens, GA 30605, USA
| | | | | |
Collapse
|
43
|
Chen P, McGuire JK, Hackman RC, Kim KH, Black RA, Poindexter K, Yan W, Liu P, Chen AJ, Parks WC, Madtes DK. Tissue inhibitor of metalloproteinase-1 moderates airway re-epithelialization by regulating matrilysin activity. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1256-70. [PMID: 18385523 DOI: 10.2353/ajpath.2008.070891] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Obliterative bronchiolitis (OB) is the histopathological finding in chronic lung allograft rejection. Mounting evidence suggests that epithelial damage drives the development of airway fibrosis in OB. Tissue inhibitor of metalloproteinase (TIMP)-1 expression increases in lung allografts and is associated with the onset of allograft rejection. Furthermore, in a mouse model of OB, airway obliteration is reduced in TIMP-1-deficient mice. Matrilysin (matrix metallproteinase-7) is essential for airway epithelial repair and is required for the re-epithelialization of airway wounds by facilitating cell migration; therefore, the goal of this study was to determine whether TIMP-1 inhibits re-epithelialization through matrilysin. We found that TIMP-1 and matrilysin co-localized in the epithelium of human lungs with OB and both co-localized and co-immunoprecipitated in wounded primary airway epithelial cultures. TIMP-1-deficient cultures migrated faster, and epithelial cells spread to a greater extent compared with wild-type cultures. TIMP-1 also inhibited matrilysin-mediated cell migration and spreading in vitro. In vivo, TIMP-1 deficiency enhanced airway re-epithelialization after naphthalene injury. Furthermore, TIMP-1 and matrilysin co-localized in airway epithelial cells adjacent to the wound edge. Our data demonstrate that TIMP-1 interacts with matrix metalloproteinases and regulates matrilysin activity during airway epithelial repair. Furthermore, we speculate that TIMP-1 overexpression restricts airway re-epithelialization by inhibiting matrilysin activity, contributing to a stereotypic injury response that promotes airway fibrosis via bronchiole airway epithelial damage and obliteration.
Collapse
Affiliation(s)
- Peter Chen
- Center for Lung Biology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Atkinson JJ, Adair-Kirk TL, Kelley DG, Demello D, Senior RM. Clara cell adhesion and migration to extracellular matrix. Respir Res 2008; 9:1. [PMID: 18179694 PMCID: PMC2249579 DOI: 10.1186/1465-9921-9-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Accepted: 01/07/2008] [Indexed: 11/10/2022] Open
Abstract
Background Clara cells are the epithelial progenitor cell of the small airways, a location known to be important in many lung disorders. Although migration of alveolar type II and bronchiolar ciliated epithelial cells has been examined, the migratory response of Clara cells has received little attention. Methods Using a modification of existing procedures for Clara cell isolation, we examined mouse Clara cells and a mouse Clara-like cell line (C22) for adhesion to and migration toward matrix substrate gradients, to establish the nature and integrin dependence of migration in Clara cells. Results We observed that Clara cells adhere preferentially to fibronectin (Fn) and type I collagen (Col I) similar to previous reports. Migration of Clara cells can be directed by a fixed gradient of matrix substrates (haptotaxis). Migration of the C22 cell line was similar to the Clara cells so integrin dependence of migration was evaluated with this cell line. As determined by competition with an RGD containing-peptide, migration of C22 cells toward Fn and laminin (Lm) 511 (formerly laminin 10) was significantly RGD integrin dependent, but migration toward Col I was RGD integrin independent, suggesting that Clara cells utilize different receptors for these different matrices. Conclusion Thus, Clara cells resemble alveolar type II and bronchiolar ciliated epithelial cells by showing integrin mediated pro-migratory changes to extracellular matrix components that are present in tissues after injury.
Collapse
Affiliation(s)
- Jeffrey J Atkinson
- Department of Internal Medicine, Pulmonary and Critical Care Division, Washington University School of Medicine, St, Louis, MO, USA.
| | | | | | | | | |
Collapse
|
45
|
Acoustically detectable cellular-level lung injury induced by fluid mechanical stresses in microfluidic airway systems. Proc Natl Acad Sci U S A 2007; 104:18886-91. [PMID: 18006663 DOI: 10.1073/pnas.0610868104] [Citation(s) in RCA: 319] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe a microfabricated airway system integrated with computerized air-liquid two-phase microfluidics that enables on-chip engineering of human airway epithelia and precise reproduction of physiologic or pathologic liquid plug flows found in the respiratory system. Using this device, we demonstrate cellular-level lung injury under flow conditions that cause symptoms characteristic of a wide range of pulmonary diseases. Specifically, propagation and rupture of liquid plugs that simulate surfactant-deficient reopening of closed airways lead to significant injury of small airway epithelial cells by generating deleterious fluid mechanical stresses. We also show that the explosive pressure waves produced by plug rupture enable detection of the mechanical cellular injury as crackling sounds.
Collapse
|
46
|
Antunes MB, Woodworth BA, Bhargave G, Xiong G, Aguilar JL, Ratner AJ, Kreindler JL, Rubenstein RC, Cohen NA. Murine nasal septa for respiratory epithelial air-liquid interface cultures. Biotechniques 2007; 43:195-6, 198, 200 passim. [PMID: 17824387 DOI: 10.2144/000112531] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Air-liquid interface models using murine tracheal respiratory epithelium have revolutionized the in vitro study of pulmonary diseases. This model is often impractical because of the small number of respiratory epithelial cells that can be isolated from the mouse trachea. We describe a simple technique to harvest the murine nasal septum and grow the epithelial cells in an air-liquid interface. The degree of ciliation of mouse trachea, nasal septum, and their respective cultured epithelium at an air-liquid interface were compared by scanning electron microscopy (SEM). Immunocytochemistry for type IV beta-tubulin and zona occludens-1 (Zo-1) are performed to determine differentiation and confluence, respectively. To rule out contamination with olfactory epithelium (OE), immunocytochemistry for olfactory marker protein (OMP) was performed. Transepithelial resistance and potential measurements were determined using a modified vertical Ussing chamber SEM reveals approximately 90% ciliated respiratory epithelium in the nasal septum as compared with 35% in the mouse trachea. The septal air-liquid interface culture demonstrates comparable ciliated respiratory epithelium to the nasal septum. Immunocytochemistry demonstrates an intact monolayer and diffuse differentiated ciliated epithelium. These cultures exhibit a transepithelial resistance and potential confirming a confluent monolayer with electrically active airway epitheliumn containing both a sodium-absorptive pathway and a chloride-secretory pathway. To increase the yield of respiratory epithelial cells harvested from mice, we have found the nasal septum is a superior source when compared with the trachea. The nasal septum increases the yield of respiratory epithelial cells up to 8-fold.
Collapse
|
47
|
Individual matrix metalloproteinases control distinct transcriptional responses in airway epithelial cells infected with Pseudomonas aeruginosa. Infect Immun 2007; 75:5640-50. [PMID: 17923522 DOI: 10.1128/iai.00799-07] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Airway epithelium is the initial point of host-pathogen interaction in Pseudomonas aeruginosa infection, an important pathogen in cystic fibrosis and nosocomial pneumonia. We used global gene expression analysis to determine airway epithelial transcriptional responses dependent on matrilysin (matrix metalloproteinase 7 [MMP-7]) and stromelysin-2 (MMP-10), two MMPs induced by acute P. aeruginosa pulmonary infection. Extraction of differential gene expression (EDGE) analysis of gene expression changes in P. aeruginosa-infected organotypic tracheal epithelial cell cultures from wild-type, Mmp7-/-, and Mmp10-/- mice identified 2,091 matrilysin-dependent and 1,628 stromelysin-2-dependent genes that were differentially expressed. Key node network analysis showed that these MMPs controlled distinct gene expression programs involved in proliferation, cell death, immune responses, and signal transduction, among other host defense processes. Our results demonstrate discrete roles for these MMPs in regulating epithelial responses to Pseudomonas infection and show that a global genomics strategy can be used to assess MMP function.
Collapse
|
48
|
Liu X, Luo M, Guo C, Yan Z, Wang Y, Engelhardt JF. Comparative biology of rAAV transduction in ferret, pig and human airway epithelia. Gene Ther 2007; 14:1543-8. [PMID: 17728794 PMCID: PMC2121584 DOI: 10.1038/sj.gt.3303014] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Differences between rodent and human airway cell biology have made it difficult to translate recombinant adeno-associated virus (rAAV)-mediated gene therapies to the lung for cystic fibrosis (CF). As new ferret and pig models for CF become available, knowledge about host cell/vector interactions in these species will become increasingly important for testing potential gene therapies. To this end, we have compared the transduction biology of three rAAV serotypes (AAV1, 2 and 5) in human, ferret, pig and mouse-polarized airway epithelia. Our results indicate that apical transduction of ferret and pig airway epithelia with these rAAV serotypes closely mirrors that observed in human epithelia (rAAV1>rAAV2 congruent withrAAV5), while transduction of mouse epithelia was significantly different (rAAV1>rAAV5>>rAAV2). Similarly, ferret, pig and human epithelia also shared serotype-specific differences in the polarity (apical vs basolateral) and proteasome dependence of rAAV transduction. Despite these parallels, N-linked sialic acid receptors were required for rAAV1 and rAAV5 transduction of human and mouse airway epithelia, but not ferret or pig airway epithelia. Hence, although the airway tropisms of rAAV serotypes 1, 2 and 5 are conserved better among ferret, pig and human as compared to mouse, viral receptors/co-receptors appear to maintain considerable species diversity.
Collapse
Affiliation(s)
- X Liu
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - M Luo
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - C Guo
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
| | - Z Yan
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Y Wang
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
| | - JF Engelhardt
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, IA, USA
- College of Life Science, Ningxia University, Yingchuan, Ningxia, China
- Department of Internal Medicine, College of Medicine, The University of Iowa, Iowa City, IA, USA
- Center for Gene Therapy of Cystic Fibrosis and Other Genetic Diseases, College of Medicine, The University of Iowa, Iowa City, IA, USA
| |
Collapse
|
49
|
Hosokawa T, Betsuyaku T, Nishimura M, Furuyama A, Katagiri K, Mochitate K. Differentiation of tracheal basal cells to ciliated cells and tissue reconstruction on the synthesized basement membrane substratum in vitro. Connect Tissue Res 2007; 48:9-18. [PMID: 17364662 DOI: 10.1080/03008200601017488] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Although lung epithelial cells directly attach to the basement membrane underneath in vivo, harvested epithelial cells are typically cultured on type I collagen gel (Col I-gel) in vitro. Recently we developed new culture substratum, designated as "synthesized Basement Membrane" (sBM), that has bared lamina densa on fibrillar collagen. To validate the usefulness of sBM substratum in airway tissue reconstitution in vitro, we cultured rat tracheal epithelial cells on sBM substratum and Col I-gel. When starting the air-liquid interface culture, most of the epithelial cells were squamous and positive for the basal cell marker cytokeratin 14 (CK14). After 14 days on sBM substratum, CK14-positive cells differentiated not only to Clara and mucous cells, but also to ciliated cells. Those differentiated cells formed pseudostratified-like epithelium and the remaining CK14-positive cells were polarized to the basal side. However, on Col I-gel, the CK14-positive cells were still squamous and not polarized, and ciliated cells did not appear. In conclusion, we established a new culture model on sBM substratum in which basal cells could differentiate to ciliated cells. The application of sBM substratum is useful in the study of the airway epithelial cell differentiation in vitro.
Collapse
Affiliation(s)
- Takeshi Hosokawa
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
MacPherson H, Keir PA, Edwards CJ, Webb S, Dorin JR. Following damage, the majority of bone marrow-derived airway cells express an epithelial marker. Respir Res 2006; 7:145. [PMID: 17177981 PMCID: PMC1764737 DOI: 10.1186/1465-9921-7-145] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 12/19/2006] [Indexed: 11/10/2022] Open
Abstract
Background Adult-derived bone marrow stem cells are capable of reconstituting the haematopoietic system. However there is ongoing debate in the literature as to whether bone marrow derived cells have the ability to populate other tissues and express tissue specific markers. The airway has been an organ of major interest and was one of the first where this was demonstrated. We have previously demonstrated that the mouse airway can be repopulated by side population bone marrow transplanted cells. Here we investigate the frequency and phenotypic nature of these bone marrow derived cells. Methods Female mice were engrafted with male whole bone marrow or side population (SP) cells and subjected to detergent-induced damage after 3 months. Donor cells were identified by Y chromosome fluorescence in situ hybridisation and their phenotype was assessed by immunohistochemistry on the same sections. Slides were visualised by a combination of widefield and deconvolved microscopy and whole cells were analysed on cytospin preparations. Results The frequencies of engraftment of male cells in the airway of mice that show this (9/10), range from 1.0 – 1.6% with whole marrow and 0.6 – 1.5% with SP cells. Undamaged controls have only between 0.1 and 0.2% male cells in the trachea. By widefield microscopy analysis we find 60.2% (53/88) of male donor derived cells express cytokeratins as a marker of epithelial cells. These results were reinforced using deconvolved microscopy and scored by two independent investigators. In addition cytospin analysis of cells dissociated from the damaged trachea of engrafted mice also reveals donor derived Y chromosome positive cells that are immunopositive for cytokeratin. Using cytokeratin and the universal haematopoietic marker CD45 immunohistochemistry, we find the donor derived cells fall into four phenotypic classes. We do not detect cytokeratin positive cells in whole bone marrow using cytokeratin immunostaining and we do not detect any cytokeratin mRNA in SP or bone marrow samples by RT-PCR. Conclusion The appearance of bone marrow derived cells in the tracheal epithelium is enriched by detergent-induced tissue damage and the majority of these cells express an epithelial marker. The cytokeratin positive donor derived cells in the tracheal epithelium are not present in the injected donor cells and must have acquired this novel phenotype in vivo.
Collapse
Affiliation(s)
- Heather MacPherson
- MRC Human Genetics Unit, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Pamela A Keir
- MRC Human Genetics Unit, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Carol J Edwards
- MRC Human Genetics Unit, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Sheila Webb
- MRC Human Genetics Unit, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| | - Julia R Dorin
- MRC Human Genetics Unit, Western General Hospital, Crewe Road South, Edinburgh EH4 2XU, UK
| |
Collapse
|