1
|
Togami K, Wolf W, Olson LC, Card M, Shen L, Schaefer A, Okuda K, Zeitlin L, Pauly M, Whaley K, Pickles RJ, Lai SK. Impact of mAb-FcRn affinity on IgG transcytosis across human well-differentiated airway epithelium. Front Immunol 2024; 15:1371156. [PMID: 39351230 PMCID: PMC11439726 DOI: 10.3389/fimmu.2024.1371156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/05/2024] [Indexed: 10/04/2024] Open
Abstract
Effective treatment and immunoprophylaxis of viral respiratory infections with neutralizing monoclonal antibodies (mAbs) require maintaining inhibitory concentrations of mAbs at the airway surface. While engineered mAbs with increased affinity to the neonatal Fc receptor (FcRn) are increasingly employed, little is known how increased affinity of Fc to FcRn influences basal-to-apical transepithelial transport (transcytosis) of mAbs across the airway epithelium. To investigate this, we utilized a model of well-differentiated human airway epithelium (WD-HAE) that exhibited robust FcRn expression, and measured the transepithelial transport of a mAb against SARS-CoV-2 Spike protein (CR3022) with either wildtype IgG1-Fc or Fc modified with YTE or LS mutations known to increase affinity for FcRn. Despite the marked differences in the affinity of these CR3022 variants for FcRn, we did not find substantial differences in basal-to-apical transport reflective of systemic dosing, or apical-to-basal transport reflective of inhaled dosing, compared to the transport of wildtype IgG1-Fc. These results suggest increasing FcRn affinity may only have limited influence over transcytosis rates of systemically dosed mAbs across the human airway epithelium over short time scales. Over longer time scales, the elevated circulating levels of mAbs with greater FcRn affinity, due to more effective FcRn-mediated recycling, may better resupply mAb into the respiratory tract, leading to more effective extended immunoprophylaxis.
Collapse
Affiliation(s)
- Kohei Togami
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Whitney Wolf
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lucas C Olson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Madison Card
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Limei Shen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alison Schaefer
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | | | | | - Raymond J Pickles
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology & Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology & Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
2
|
Figueiredo IAD, Ferreira SRD, Fernandes JM, Silva BA, Vasconcelos LHC, Cavalcante FA. A review of the pathophysiology and the role of ion channels on bronchial asthma. Front Pharmacol 2023; 14:1236550. [PMID: 37841931 PMCID: PMC10568497 DOI: 10.3389/fphar.2023.1236550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Asthma is one of the main non-communicable chronic diseases and affects a huge portion of the population. It is a multifactorial disease, classified into several phenotypes, being the allergic the most frequent. The pathophysiological mechanism of asthma involves a Th2-type immune response, with high concentrations of allergen-specific immunoglobulin E, eosinophilia, hyperreactivity and airway remodeling. These mechanisms are orchestrated by intracellular signaling from effector cells, such as lymphocytes and eosinophils. Ion channels play a fundamental role in maintaining the inflammatory response on asthma. In particular, transient receptor potential (TRP), stock-operated Ca2+ channels (SOCs), Ca2+-activated K+ channels (IKCa and BKCa), calcium-activated chloride channel (TMEM16A), cystic fibrosis transmembrane conductance regulator (CFTR), piezo-type mechanosensitive ion channel component 1 (PIEZO1) and purinergic P2X receptor (P2X). The recognition of the participation of these channels in the pathological process of asthma is important, as they become pharmacological targets for the discovery of new drugs and/or pharmacological tools that effectively help the pharmacotherapeutic follow-up of this disease, as well as the more specific mechanisms involved in worsening asthma.
Collapse
Affiliation(s)
- Indyra Alencar Duarte Figueiredo
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Sarah Rebeca Dantas Ferreira
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Jayne Muniz Fernandes
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Luiz Henrique César Vasconcelos
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Fabiana de Andrade Cavalcante
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| |
Collapse
|
3
|
Martinez Ledo A, Thibodeaux S, Duong L, Altinoglu E, Dimke T, Shaw D, Rowlands D, Growcott E. Aerosol technology to mimic dry powder inhalation in vitro using pulmonary cell models. Eur J Pharm Biopharm 2023:S0939-6411(23)00123-6. [PMID: 37196872 DOI: 10.1016/j.ejpb.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/21/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Inhaled therapy confers key advantages for the treatment of topical pulmonary diseases and offers potential for systemic delivery of medicines. Dry powder inhalers (DPIs) are generally the preferred devices for pulmonary delivery due to improved stability and satisfactory patient compliance. However, the mechanisms governing drug powder dissolution and availability in the lung and poorly understood. Here, we report a new in vitro system to study epithelial absorption of inhaled dry powders in lung barrier models of the upper and lower airway. The system is based on a CULTEX® RFS (Radial Flow System) cell exposure module joined to a Vilnius aerosol generator and allows the coupling of drug dissolution and permeability assessments. The cellular models recapitulate the barrier morphology and function of healthy and diseased pulmonary epithelium and incorporate the mucosal barrier to enable the investigation of drug powder dissolution in biorelevant conditions. With this system, we found differences in permeability across the airway tree and pinpointed the impact of diseased barriers in paracellular drug transport. Furthermore, we identified a different rank order of permeability for compounds tested in solution or powder form. These results highlight the value of this in vitro drug aerosolization setup for use in research and development of inhaled medicines.
Collapse
Affiliation(s)
- Adriana Martinez Ledo
- Disease Area X, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States
| | - Stefan Thibodeaux
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States
| | - Lisa Duong
- Oncology, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States
| | - Erhan Altinoglu
- Chemical and Pharmaceutical Profiling, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States
| | - Thomas Dimke
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Duncan Shaw
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States
| | - David Rowlands
- Disease Area X, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States.
| | - Ellena Growcott
- Disease Area X, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, 02139, United States.
| |
Collapse
|
4
|
Cognetti JS, Moen MT, Brewer MG, Bryan MR, Tice JD, McGrath JL, Miller BL. A photonic biosensor-integrated tissue chip platform for real-time sensing of lung epithelial inflammatory markers. LAB ON A CHIP 2023; 23:239-250. [PMID: 36594179 PMCID: PMC10311125 DOI: 10.1039/d2lc00864e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Tissue chip (TC) devices, also known as microphysiological systems (MPS) or organ chips (OCs or OoCs), seek to mimic human physiology on a small scale. They are intended to improve upon animal models in terms of reproducibility and human relevance, at a lower monetary and ethical cost. Virtually all TC systems are analyzed at an endpoint, leading to widespread recognition that new methods are needed to enable sensing of specific biomolecules in real time, as they are being produced by the cells. To address this need, we incorporated photonic biosensors for inflammatory cytokines into a model TC. Human bronchial epithelial cells seeded in a microfluidic device were stimulated with lipopolysaccharide, and the cytokines secreted in response sensed in real time. Sensing analyte transport through the TC in response to disruption of tissue barrier was also demonstrated. This work demonstrates the first application of photonic sensors to a human TC device, and will enable new applications in drug development and disease modeling.
Collapse
Affiliation(s)
- John S Cognetti
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
| | - Maya T Moen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
| | - Matthew G Brewer
- Department of Dermatology, University of Rochester, Rochester, NY 14642, USA
| | - Michael R Bryan
- Department of Dermatology, University of Rochester, Rochester, NY 14642, USA
| | | | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Program in Materials Science, University of Rochester, Rochester, NY 14642, USA
| | - Benjamin L Miller
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Department of Dermatology, University of Rochester, Rochester, NY 14642, USA
- Program in Materials Science, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
5
|
Rothen-Rutishauser B, Gibb M, He R, Petri-Fink A, Sayes CM. Human lung cell models to study aerosol delivery - considerations for model design and development. Eur J Pharm Sci 2023; 180:106337. [PMID: 36410570 DOI: 10.1016/j.ejps.2022.106337] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Human lung tissue models range from simple monolayer cultures to more advanced three-dimensional co-cultures. Each model system can address the interactions of different types of aerosols and the choice of the model and the mode of aerosol exposure depends on the relevant scenario, such as adverse outcomes and endpoints of interest. This review focuses on the functional, as well as structural, aspects of lung tissue from the upper airway to the distal alveolar compartments as this information is relevant for the design of a model as well as how the aerosol properties determine the interfacial properties with the respiratory wall. The most important aspects on how to design lung models are summarized with a focus on (i) choice of appropriate scaffold, (ii) selection of cell types for healthy and diseased lung models, (iii) use of culture condition and assembly, (iv) aerosol exposure methods, and (v) endpoints and verification process. Finally, remaining challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Barbara Rothen-Rutishauser
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland.
| | - Matthew Gibb
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA
| | - Ruiwen He
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA.
| |
Collapse
|
6
|
Di Cristo L, Sabella S. Cell Cultures at the Air-Liquid Interface and Their Application in Cancer Research. Methods Mol Biol 2023; 2645:41-64. [PMID: 37202611 DOI: 10.1007/978-1-0716-3056-3_2] [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] [Indexed: 05/20/2023]
Abstract
Air-liquid interface (ALI) cell cultures are considered a valid tool for the replacement of animals in biomedical research. By mimicking crucial features of the human in vivo epithelial barriers (e.g., lung, intestine, and skin), ALI cell cultures enable proper structural architectures and differentiated functions of normal and diseased tissue barriers. Thereby, ALI models realistically resemble tissue conditions and provide in vivo-like responses. Since their implementation, they are routinely used in several applications, from toxicity testing to cancer research, receiving an appreciable level of acceptance (in some cases a regulatory acceptance) as attractive testing alternatives to animals. In this chapter, an overview of the ALI cell cultures will be presented together with their application in cancer cell culture, highlighting the potential advantages and disadvantages of the model.
Collapse
Affiliation(s)
- Luisana Di Cristo
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy.
| | - Stefania Sabella
- D3 PharmaChemistry, Nanoregulatory Group, Italian Institute of Technology, Genoa, Italy
| |
Collapse
|
7
|
Yang S, Cheng Y, Chen Z, Liu T, Yin L, Pu Y, Liang G. In vitro evaluation of nanoplastics using human lung epithelial cells, microarray analysis and co-culture model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112837. [PMID: 34619472 DOI: 10.1016/j.ecoenv.2021.112837] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 05/22/2023]
Abstract
Nanoplastics, including polystyrene nanoplastics (PS-NPs), are widely existed in the atmosphere, which can be directly and continuously inhaled into the human body, posing a serious threat to the respiratory system. Therefore, it is urgent to estimate the potential pulmonary toxicity of airborne NPs and understand its underlying mechanism. In this research, we used two types of human lung epithelial cells (bronchial epithelium transformed with Ad12-SV40 2B, BEAS-2B) and (human pulmonary alveolar epithelial cells, HPAEpiC) to investigate the association between lung injury and PS-NPs. We found PS-NPs could significantly reduce cell viability in a dose-dependent manner and selected 7.5, 15 and 30 μg/cm2 PS-NPs as the exposure dosage levels. Microarray detection revealed that 770 genes in the 7.5 μg/cm2 group and 1951 genes in the 30 μg/cm2 group were distinctly altered compared to the control group. Function analysis suggested that redox imbalance might play central roles in PS-NPs induced lung injury. Further experiments verified that PS-NPs could break redox equilibrium, induce inflammatory effects, and triggered apoptotic pathways to cause cell death. Importantly, we found that PS-NPs could decrease transepithelial electrical resistance by depleting tight junctional proteins. Result also demonstrated that PS-NPs-treated cells increased matrix metallopeptidase 9 and Surfactant protein A levels, suggesting the exposure of PS-NPs might reduce the repair ability of the lung and cause tissue damage. In conclusion, nanoplastics could induce oxidative stress and inflammatory responses, followed by cell death and epithelial barrier destruction, which might result in tissue damage and lung disease after prolonged exposure.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| | - Yanping Cheng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, Jiangsu, PR China.
| | - Tong Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, PR China.
| |
Collapse
|
8
|
Qin W, Brands X, van’t Veer C, F. de Vos A, Sirard JC, J. T. H. Roelofs J, P. Scicluna B, van der Poll T. Bronchial epithelial DNA methyltransferase 3b dampens pulmonary immune responses during Pseudomonas aeruginosa infection. PLoS Pathog 2021; 17:e1009491. [PMID: 33793661 PMCID: PMC8043394 DOI: 10.1371/journal.ppat.1009491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/13/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023] Open
Abstract
DNA methyltransferase (Dnmt)3b mediates de novo DNA methylation and modulation of Dnmt3b in respiratory epithelial cells has been shown to affect the expression of multiple genes. Respiratory epithelial cells provide a first line of defense against pulmonary pathogens and play a crucial role in the immune response during pneumonia caused by Pseudomonas (P.) aeruginosa, a gram-negative bacterium that expresses flagellin as an important virulence factor. We here sought to determine the role of Dntm3b in respiratory epithelial cells in immune responses elicited by P. aeruginosa. DNMT3B expression was reduced in human bronchial epithelial (BEAS-2B) cells as well as in primary human and mouse bronchial epithelial cells grown in air liquid interface upon exposure to P. aeruginosa (PAK). Dnmt3b deficient human bronchial epithelial (BEAS-2B) cells produced more CXCL1, CXCL8 and CCL20 than control cells when stimulated with PAK, flagellin-deficient PAK (PAKflic) or flagellin. Dnmt3b deficiency reduced DNA methylation at exon 1 of CXCL1 and enhanced NF-ĸB p65 binding to the CXCL1 promoter. Mice with bronchial epithelial Dntm3b deficiency showed increased Cxcl1 mRNA expression in bronchial epithelium and CXCL1 protein release in the airways during pneumonia caused by PAK, which was associated with enhanced neutrophil recruitment and accelerated bacterial clearance; bronchial epithelial Dnmt3b deficiency did not modify responses during pneumonia caused by PAKflic or Klebsiella pneumoniae (an un-flagellated gram-negative bacterium). Dnmt3b deficiency in type II alveolar epithelial cells did not affect mouse pulmonary defense against PAK infection. These results suggest that bronchial epithelial Dnmt3b impairs host defense during Pseudomonas induced pneumonia, at least in part, by dampening mucosal responses to flagellin.
Collapse
Affiliation(s)
- Wanhai Qin
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Xanthe Brands
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cornelis van’t Veer
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex F. de Vos
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Claude Sirard
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Joris J. T. H. Roelofs
- Department of Pathology, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Brendon P. Scicluna
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Zhou J, Zhou XD, Xu R, Du XZ, Li Q, Li B, Zhang GY, Chen LX, Perelman JM, Kolosov VP. The Degradation of Airway Epithelial Tight Junctions in Asthma Under High Airway Pressure Is Probably Mediated by Piezo-1. Front Physiol 2021; 12:637790. [PMID: 33868003 PMCID: PMC8047413 DOI: 10.3389/fphys.2021.637790] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/02/2022] Open
Abstract
Full functioning of the airway physical barrier depends on cellular integrity, which is coordinated by a series of tight junction (TJ) proteins. Due to airway spasm, edema, and mucus obstruction, positive end-expiratory alveolar pressure (also termed auto-PEEP) is a common pathophysiological phenomenon, especially in acute asthma attack. However, the influence of auto-PEEP on small airway epithelial TJs is currently unclear. We performed studies to investigate the effect of extra pressure on small airway epithelial TJs and its mechanism. The results first confirmed that a novel mechanosensitive receptor, piezo-1, was highly expressed in the airway epithelium of asthmatic mice. Extra pressure induced the degradation of occludin, ZO-1 and claudin-18 in primary human small airway epithelial cells (HSAECs), resulting in a decrease in transepithelial electrical resistance (TER) and an increase in cell layer permeability. Through in vitro investigations, we observed that exogenous pressure stimulation could elevate the intracellular calcium concentration ([Ca2+]i) in HSAECs. Downregulation of piezo-1 with siRNA and pretreatment with BAPTA-AM or ALLN reduced the degradation of TJs and attenuated the impairment of TJ function induced by exogenous pressure. These findings indicate the critical role of piezo-1/[Ca2+]i/calpain signaling in the regulation of small airway TJs under extra pressure stimulation.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xian-Zhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Bin Li
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fengjie, Chongqing, China
| | - Guo-Yue Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Xiu Chen
- Department of Respiratory Medicine, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Juliy M Perelman
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| | - Victor P Kolosov
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| |
Collapse
|
10
|
Fergie N, Todd N, McClements L, McAuley D, O’Kane C, Krasnodembskaya A. Hypercapnic acidosis induces mitochondrial dysfunction and impairs the ability of mesenchymal stem cells to promote distal lung epithelial repair. FASEB J 2019; 33:5585-5598. [PMID: 30649987 PMCID: PMC6436662 DOI: 10.1096/fj.201802056r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 01/02/2019] [Indexed: 01/27/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating disorder characterized by diffuse inflammation and edema formation. The main management strategy, low tidal volume ventilation, can be associated with the development of hypercapnic acidosis (HCA). Mesenchymal stem cells (MSCs) are a promising therapeutic candidate currently in early-phase clinical trials. The effects of HCA on the alveolar epithelium and capillary endothelium are not well established. The therapeutic efficacy of MSCs has never been reported in HCA. In the present study, we evaluated the effects of HCA on inflammatory response and reparative potential of the primary human small airway epithelial and lung microvasculature endothelial cells as well as on the capacity of bone marrow-derived MSCs to promote wound healing in vitro. We demonstrate that HCA attenuates the inflammatory response and reparative potential of primary human small airway epithelium and capillary endothelium and induces mitochondrial dysfunction. It was found that MSCs promote lung epithelial wound repair via the transfer of functional mitochondria; however, this proreparative effect of MSCs was lost in the setting of HCA. Therefore, HCA may adversely impact recovery from ARDS at the cellular level, whereas MSCs may not be therapeutically beneficial in patients with ARDS who develop HCA.-Fergie, N., Todd, N., McClements, L., McAuley, D., O'Kane, C., Krasnodembskaya, A. Hypercapnic acidosis induces mitochondrial dysfunction and impairs the ability of mesenchymal stem cells to promote distal lung epithelial repair.
Collapse
Affiliation(s)
- Nicola Fergie
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| | - Naomi Todd
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| | - Lana McClements
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| | - Danny McAuley
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| | - Cecilia O’Kane
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| | - Anna Krasnodembskaya
- Centre for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University of Belfast, Belfast, United Kingdom
| |
Collapse
|
11
|
Alekseenko SI, Skalny AV, Ajsuvakova OP, Skalnaya MG, Notova SV, Tinkov AA. Mucociliary transport as a link between chronic rhinosinusitis and trace element dysbalance. Med Hypotheses 2019; 127:5-10. [PMID: 31088648 DOI: 10.1016/j.mehy.2019.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/01/2019] [Accepted: 03/20/2019] [Indexed: 12/21/2022]
Abstract
Chronis rhinosinusitis is considered as a widespread public health issue with a prevalence of 10%. The disease significantly reduces quality of life and increases the risk of cardiovascular diseases as well as certain forms of cancer. Alteration of mucociliary clearance frequently observed in the patients and plays a significant role in disease pathogenesis. Certain studies have demonstrated that patients with chronic rhinosinusitis are characterized by significant reduction of essential trace elements and toxic metal overload. However, the particular mechanisms of the role of trace element dysbalance in chronic rhinosinusitis are unclear. We hypothesize that exposure to toxic trace elements (arsenic, nickel, cadmium) damages ciliary mucosal epithelium thus affecting mucociliary transport. In turn, altered mucociliary transport results in reduced removal of the inhaled metal-containing particles from nasal mucosa leading to their absorption and further aggravation of toxicity. Essential trace elements (zinc, selenium) play a significant role in regulation of mucociliary transport and immunity, thus their deficiency (either dietary or due to antagonism with toxic metals) may be associated with impaired functions and increased toxic metal toxicity. Therefore, a vicious circle involving metal accumulation and toxicity, essential element deficiency, impairment of mucociliary transport and metal particle removal, resulting in further accumulation of metals and aggravation of toxic effects is formed. The present hypothesis is supported by the findings on the impact of trace elements especially zinc and arsenic on mucociliary clearance, the role of mucociliary transport in heavy metal particles elimination from the airways, trace element dysbalance in chronic rhinosinusitis, as well as toxic and essential metal antagonism. The data from hypothesis testing and its verification may be used for development of therapeutic approach for management of chronic rhinosinusitis. Particularly, the use of essential elements (zinc, selenium) may reduce toxic metal toxicity thus destroying the vicious circle of heavy metal exposure, toxicity, alteration of mucociliary clearance, and aggravation of chronic rhinosinusitis. Essential element supplementation may be considered as a tool for management of chronic refractory rhinosinusitis. In addition, analysis of essential and toxic trace element status may provide an additional diagnostic approach to risk assessment of chronic rhinosinusitis in highly polluted environments.
Collapse
Affiliation(s)
- Svetlana I Alekseenko
- K. A. Raukhfuss Children's Municipal Multidisciplinary Clinical Center of High Medical Technologies, St. Petersburg, Russia; Mechnikov North-West State Medical University, St Petersburg, Russia
| | - Anatoly V Skalny
- Yaroslavl State University, Yaroslavl, Russia; Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olga P Ajsuvakova
- Yaroslavl State University, Yaroslavl, Russia; Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Margarita G Skalnaya
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Svetlana V Notova
- Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia; Orenburg State University, Orenburg, Russia
| | - Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia; Peoples' Friendship University of Russia (RUDN University), Moscow, Russia; I.M. Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
12
|
Changes in miRNA Gene Expression during Wound Repair in Differentiated Normal Human Bronchial Epithelium. Int J Genomics 2018; 2018:9093785. [PMID: 30255030 PMCID: PMC6145058 DOI: 10.1155/2018/9093785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/12/2018] [Accepted: 08/08/2018] [Indexed: 11/18/2022] Open
Abstract
Purpose Airway epithelium acts as a protective barrier against the particles from the inhaled air. Damage to the epithelium may result in loss of the barrier function. Epithelial repair in response to injury requires complex mechanisms, such as microRNA, small noncoding molecules, to regulate the processes involved in wound repair. We aimed to establish if the microRNA gene expression profile is altered during the airway epithelial repair in differentiated cells. Methods miRNA gene expression profile during the wound closure of differentiated normal human bronchial epithelium (NHBE) from one donor was analysed using quantitative real-time PCR. We have analysed the expression of 754 genes at five time points during a 48-hour period of epithelium repair using TaqMan Low Density Array. Results We found out that 233 miRNA genes were expressed in normal human bronchial epithelium. Twenty miRNAs were differentially expressed during the wound repair process, but only one (miR-455-3p) showed significance after FDR adjustment (p = 0.02). Using STEM, we have identified two clusters of several miRNA genes with similar expression profile. Pathway enrichment analysis showed several significant signaling pathways altered during repair, mainly involved in cell cycle regulation, proliferation, migration, adhesion, and transcription regulation. Conclusions miRNA expression profile is altered during airway epithelial repair of differentiated cells from one donor in response to mechanical injury in vitro, suggesting their potential role in wound repair.
Collapse
|
13
|
Lee H, Lee J, Hong SH, Rahman I, Yang SR. Inhibition of RAGE Attenuates Cigarette Smoke-Induced Lung Epithelial Cell Damage via RAGE-Mediated Nrf2/DAMP Signaling. Front Pharmacol 2018; 9:684. [PMID: 30013476 PMCID: PMC6036614 DOI: 10.3389/fphar.2018.00684] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
The oxidative stress and cellular apoptosis by environmental factor including cigarette smoke induces alveolar airway remodeling leading to chronic obstructive pulmonary disease (COPD). Recently, the receptor for advanced glycan end products (RAGE) which is highly expressed in alveolar epithelium is emerging as a biomarker for COPD susceptibility or progression. However, it still remains unknown how RAGE plays a role in cigarette smoke extract (CSE)-exposed human alveolar type II epithelial cell line. Therefore, we determined the efficacy of RAGE-specific antagonist FPS-ZM1 in response to CSE-induced lung epithelial cells. CSE induced the elevated generation of RONS and release of pro-inflammatory cytokines, and impaired the cellular antioxidant defense system. Further, CSE induced the alteration of RAGE distribution via the activation of redox-sensitive DAMP (Damage-associated molecular patterns) signaling through Nrf2 in cells. Although pre-treatment with SB202190 (p38 inhibitor) or SP600125 (JNK inhibitor) failed to recover the alteration of RAGE distribution, treatment of FPS-ZM1 significantly exhibited anti-inflammatory and anti-oxidative/nitrosative effects, also inhibited the activation of redox-sensitive DAMP signaling through Nrf2 (nuclear factor erythroid 2-related factor 2) migration in the presence of CSE. Taken together, our data demonstrate that RAGE and Nrf2 play a pivotal role in maintenance of alveolar epithelial integrity.
Collapse
Affiliation(s)
- Hanbyeol Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea
| | - Jooyeon Lee
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, South Korea
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, Kangwon National University, Chuncheon, South Korea,*Correspondence: Se-Ran Yang,
| |
Collapse
|
14
|
Tenenbaum-Katan J, Artzy-Schnirman A, Fishler R, Korin N, Sznitman J. Biomimetics of the pulmonary environment in vitro: A microfluidics perspective. BIOMICROFLUIDICS 2018; 12:042209. [PMID: 29887933 PMCID: PMC5973897 DOI: 10.1063/1.5023034] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/20/2018] [Indexed: 05/08/2023]
Abstract
The entire luminal surface of the lungs is populated with a complex yet confluent, uninterrupted airway epithelium in conjunction with an extracellular liquid lining layer that creates the air-liquid interface (ALI), a critical feature of healthy lungs. Motivated by lung disease modelling, cytotoxicity studies, and drug delivery assessments amongst other, in vitro setups have been traditionally conducted using macroscopic cultures of isolated airway cells under submerged conditions or instead using transwell inserts with permeable membranes to model the ALI architecture. Yet, such strategies continue to fall short of delivering a sufficiently realistic physiological in vitro airway environment that cohesively integrates at true-scale three essential pillars: morphological constraints (i.e., airway anatomy), physiological conditions (e.g., respiratory airflows), and biological functionality (e.g., cellular makeup). With the advent of microfluidic lung-on-chips, there have been tremendous efforts towards designing biomimetic airway models of the epithelial barrier, including the ALI, and leveraging such in vitro scaffolds as a gateway for pulmonary disease modelling and drug screening assays. Here, we review in vitro platforms mimicking the pulmonary environment and identify ongoing challenges in reconstituting accurate biological airway barriers that still widely prevent microfluidic systems from delivering mainstream assays for the end-user, as compared to macroscale in vitro cell cultures. We further discuss existing hurdles in scaling up current lung-on-chip designs, from single airway models to more physiologically realistic airway environments that are anticipated to deliver increasingly meaningful whole-organ functions, with an outlook on translational and precision medicine.
Collapse
Affiliation(s)
- Janna Tenenbaum-Katan
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Rami Fishler
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| |
Collapse
|
15
|
Bhowmick R, Derakhshan T, Liang Y, Ritchey J, Liu L, Gappa-Fahlenkamp H. A Three-Dimensional Human Tissue-Engineered Lung Model to Study Influenza A Infection. Tissue Eng Part A 2018; 24:1468-1480. [PMID: 29732955 DOI: 10.1089/ten.tea.2017.0449] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) claims ∼250,000-500,000 lives annually worldwide. Currently, there are a few in vitro models available to study IAV immunopathology. Monolayer cultures of cell lines and primary lung cells (two-dimensional [2D] cell culture) is the most commonly used tool, however, this system does not have the in vivo-like structure of the lung and immune responses to IAV as it lacks the three-dimensional (3D) tissue structure. To recapitulate the lung physiology in vitro, a system that contains multiple cell types within a 3D environment that allows cell movement and interaction would provide a critical tool. In this study, as a first step in designing a 3D-Human Tissue-Engineered Lung Model (3D-HTLM), we describe the 3D culture of primary human small airway epithelial cells (HSAEpCs) and determined the immunophenotype of this system in response to IAV infections. We constructed a 3D chitosan-collagen scaffold and cultured HSAEpCs on these scaffolds at air-liquid interface (ALI). These 3D cultures were compared with 2D-cultured HSAEpCs for viability, morphology, marker protein expression, and cell differentiation. Results showed that the 3D-cultured HSAEpCs at ALI yielded maximum viable cells and morphologically resembled the in vivo lower airway epithelium. There were also significant increases in aquaporin-5 and cytokeratin-14 expression for HSAEpCs cultured in 3D compared to 2D. The 3D culture system was used to study the infection of HSAEpCs with two major IAV strains, H1N1 and H3N2. The HSAEpCs showed distinct changes in marker protein expression, both at mRNA and protein levels, and the release of proinflammatory cytokines. This study is the first step in the development of the 3D-HTLM, which will have wide applicability in studying pulmonary pathophysiology and therapeutics development.
Collapse
Affiliation(s)
- Rudra Bhowmick
- 1 School of Chemical Engineering, Oklahoma State University , Stillwater, Oklahoma
| | - Tahereh Derakhshan
- 1 School of Chemical Engineering, Oklahoma State University , Stillwater, Oklahoma
| | - Yurong Liang
- 2 Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | - Jerry Ritchey
- 3 Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | - Lin Liu
- 2 Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | | |
Collapse
|
16
|
Lee KW, Nam MH, Lee HR, Hong CO, Lee KW. Protective effects of chebulic acid on alveolar epithelial damage induced by urban particulate matter. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:373. [PMID: 28724416 PMCID: PMC5518117 DOI: 10.1186/s12906-017-1870-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/04/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Chebulic acid (CA) isolated from T. chebula, which has been reported for treating asthma, as a potent anti-oxidant resources. Exposure to ambient urban particulate matter (UPM) considered as a risk for cardiopulmonary vascular dysfunction. To investigate the protective effect of CA against UPM-mediated collapse of the pulmonary alveolar epithelial (PAE) cell (NCI-H441), barrier integrity parameters, and their elements were evaluated in PAE. METHODS CA was acquired from the laboratory previous reports. UPM was obtained from the National Institutes of Standards and Technology, and these were collected in St. Louis, MO, over a 24-month period and used as a standard reference. To confirm the protection of PAE barrier integrity, paracellular permeability and the junctional molecules were estimated with determination of transepithelial electrical resistance, Western Blotting, RT-PCR, and fluorescent staining. RESULTS UPM aggravated the generation of reactive oxygen species (ROS) in PAE and also decreased mRNA and protein levels of junction molecules and barrier integrity in NCI-H441. However, CA repressed the ROS in PAE, also improved barrier integrity by protecting the junctional parameters in NCI-H411. CONCLUSIONS These data showed that CA resulted in decreased UPM-induced ROS formation, and the protected the integrity of the tight junctions against UPM exposure to PAE barrier.
Collapse
|
17
|
Narożna B, Langwinski W, Jackson C, Lackie P, Holloway JW, Szczepankiewicz A. MicroRNA-328 is involved in wound repair process in human bronchial epithelial cells. Respir Physiol Neurobiol 2017; 242:59-65. [PMID: 28347890 DOI: 10.1016/j.resp.2017.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 03/06/2017] [Accepted: 03/24/2017] [Indexed: 12/26/2022]
Abstract
Our aim was to investigate the role of microRNA on epithelial wound repair by global microRNA silencing. We have also analysed the influence of five miRNAs (miR-328, miR-342, miR-411, miR-609, miR-888, previously identified) on wound repair in 16HBE14o-bronchial epithelial cell line. Cells were transfected with siRNAs against human DROSHA and DICER1 or miRNA mimics or inhibitors. Wounding assays were performed and the cells were observed using time-lapse microscopy. The area of damage was calculated at chosen time points, followed by data analysis. Cells with silenced global miRNA expression showed a significantly slower repair rate compared to the control cells (p=0.001). For miR-328, we observed significantly delayed repair in cells transfected with the inhibitor compared to control (p=0.02). Global microRNA silencing significantly decreased the repair rate of airway epithelial cells in vitro, indicating an important role of miRNA in the regulation of wound repair and that miR-328, possibly involved in actin pathway, may be a potent modifier of this process.
Collapse
Affiliation(s)
- Beata Narożna
- Laboratory of Molecular and Cell Biology, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 27/33 Szpitalna St., 60-572 Poznan, Poland
| | - Wojciech Langwinski
- Laboratory of Molecular and Cell Biology, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 27/33 Szpitalna St., 60-572 Poznan, Poland
| | - Claire Jackson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Peter Lackie
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Aleksandra Szczepankiewicz
- Laboratory of Molecular and Cell Biology, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 27/33 Szpitalna St., 60-572 Poznan, Poland.
| |
Collapse
|
18
|
Jimenez FR, Lewis JB, Belgique ST, Milner DC, Lewis AL, Dunaway TM, Egbert KM, Winden DR, Arroyo JA, Reynolds PR. Cigarette smoke and decreased oxygen tension inhibit pulmonary claudin-6 expression. Exp Lung Res 2016; 42:440-452. [PMID: 27982694 DOI: 10.1080/01902148.2016.1261309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Chronic obstructive pulmonary disease is a condition involving perturbed barrier integrity coincident with both emphysema and inflammation of the airways, and smoking is considered a major risk factor. Claudins (Cldns) stabilize barriers and contribute to tight junctions by preventing paracellular transport of extracellular fluid constituents. METHODS To determine Cldn6 was differentially influenced by tobacco smoke, Cldn6 was evaluated in cells and tissues by q-PCR, immunoblotting, and immunohistochemistry following exposure. Cldn6 transcriptional regulation was also assessed using luciferase reporter constructs. RESULTS Q-PCR and immunoblotting revealed that Cldn6 was decreased in alveolar type II-like epithelial cells (A549) and primary small airway epithelial cells when exposed to cigarette smoke extract (CSE). Cldn6 was also markedly decreased in the lungs of mice exposed to acute tobacco smoke delivered by a nose-only automated smoke machine compared to controls. Luciferase reporter assays incorporating 0.5-kb, 1.0-kb, or 2.0-kb of the Cldn6 promoter revealed decreased transcription of Cldn6 following exposure to CSE. Cldn6 transcriptional regulation was also assessed in hypoxic conditions due to low oxygen tension observed during smoking. Hypoxia and hypoxia inducible factor-1 alpha caused decreased transcription of the Cldn6 gene via interactions with putative response elements in the proximal promoter sequence. CONCLUSIONS These data reveal that tight junctional proteins such as Cldn6 are differentially regulated by tobacco-smoke exposure and that Cldns are potentially targeted when epithelial cells respond to tobacco smoke. Further research may show that Cldns expressed in tight junctions between parenchymal cells contribute to impaired structural integrity of the lung coincident with smoking.
Collapse
Affiliation(s)
- Felix R Jimenez
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Josh B Lewis
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Samuel T Belgique
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Dallin C Milner
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Adam L Lewis
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Todd M Dunaway
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Kaleb M Egbert
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Duane R Winden
- b College of Dental Medicine, Roseman University of Health Sciences-South Jordan Campus , South Jordan , Utah , USA
| | - Juan A Arroyo
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| | - Paul R Reynolds
- a Lung and Placenta Research Laboratory, Department of Physiology and Developmental Biology , Brigham Young University , Provo , Utah , USA
| |
Collapse
|
19
|
Papazian D, Würtzen PA, Hansen SWK. Polarized Airway Epithelial Models for Immunological Co-Culture Studies. Int Arch Allergy Immunol 2016; 170:1-21. [PMID: 27240620 DOI: 10.1159/000445833] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Epithelial cells line all cavities and surfaces throughout the body and play a substantial role in maintaining tissue homeostasis. Asthma and other atopic diseases are increasing worldwide and allergic disorders are hypothesized to be a consequence of a combination of dysregulation of the epithelial response towards environmental antigens and genetic susceptibility, resulting in inflammation and T cell-derived immune responses. In vivo animal models have long been used to study immune homeostasis of the airways but are limited by species restriction and lack of exposure to a natural environment of both potential allergens and microflora. Limitations of these models prompt a need to develop new human cell-based in vitro models. A variety of co-culture systems for modelling the respiratory epithelium exist and are available to the scientific community. The models have become increasingly sophisticated and specific care needs to be taken with regard to cell types, culture medium and culture models, depending on the aim of the study. Although great strides have been made, there is still a need for further optimization, and optimally also for standardization, in order for in vitro co-culture models to become powerful tools in the discovery of key molecules dictating immunity and/or tolerance, and for understanding the complex interplay that takes place between mucosa, airway epithelium and resident or infiltrating immune cells. This review focuses on current knowledge and the advantages and limitations of the different cell types and culture methods used in co-culture models of the human airways.
Collapse
Affiliation(s)
- Dick Papazian
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
20
|
Shang VCM, O'Sullivan SE, Kendall DA, Roberts RE. The endogenous cannabinoid anandamide increases human airway epithelial cell permeability through an arachidonic acid metabolite. Pharmacol Res 2016; 105:152-63. [PMID: 26808080 DOI: 10.1016/j.phrs.2016.01.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 01/18/2016] [Accepted: 01/19/2016] [Indexed: 01/31/2023]
Abstract
Injury to the bronchial epithelium in respiratory diseases such as asthma and COPD results in the loss of barrier function and an elevated sensitivity to environmental insults. An increased release of the endogenous cannabinoid, anandamide in response to inhalation of allergen in asthmatic patients has been reported. The aim of this study was, therefore, to determine the effects of endocannabinoids on bronchial epithelial cell permeability and to investigate the mechanisms involved. Calu-3 human bronchial epithelial cells were cultured at air-liquid interface to allow development of tight junctions. Changes in Transepithelial Electrical Resistance (TEER), a reflection of epithelial permeability, were measured at various time points post-treatment, and expression of the tight junction proteins, occludin and ZO-1, were determined using Western immunoblotting. Anandamide produced a significant reduction in TEER, which was unaffected by cannabinoid receptor antagonists, but attenuated by URB597, an inhibitor of fatty acid amide hydrolase, and by a combination of cyclooxygenase (COX) and lipoxygenase (LOX) blockade. The anandamide metabolite, arachidonic acid, showed similar TEER decrease that was also prevented in the presence of COX and LOX inhibitor. Expression of occludin and ZO-1 were also reduced by anandamide. These findings indicate a pro-inflammatory-like effect of anandamide on bronchial epithelial permeability, mediated by cyclooxygenase and lipoxygenase metabolites, and suggest that inhibition of anandamide degradation might provide a novel approach to treat airway inflammation.
Collapse
Affiliation(s)
- V C M Shang
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | - S E O'Sullivan
- School of Graduate Entry Medicine and Health, Royal Derby Hospital, University of Nottingham, DE22 3DT, United Kingdom
| | - D A Kendall
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - R E Roberts
- School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
21
|
Early Detection of Junctional Adhesion Molecule-1 (JAM-1) in the Circulation after Experimental and Clinical Polytrauma. Mediators Inflamm 2015; 2015:463950. [PMID: 26556956 PMCID: PMC4628652 DOI: 10.1155/2015/463950] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 01/31/2023] Open
Abstract
Severe tissue trauma-induced systemic inflammation is often accompanied by evident or occult blood-organ barrier dysfunctions, frequently leading to multiple organ dysfunction. However, it is unknown whether specific barrier molecules are shed into the circulation early after trauma as potential indicators of an initial barrier dysfunction. The release of the barrier molecule junctional adhesion molecule-1 (JAM-1) was investigated in plasma of C57BL/6 mice 2 h after experimental mono- and polytrauma as well as in polytrauma patients (ISS ≥ 18) during a 10-day period. Correlation analyses were performed to indicate a linkage between JAM-1 plasma concentrations and organ failure. JAM-1 was systemically detected after experimental trauma in mice with blunt chest trauma as a driving force. Accordingly, JAM-1 was reduced in lung tissue after pulmonary contusion and JAM-1 plasma levels significantly correlated with increased protein levels in the bronchoalveolar lavage as a sign for alveolocapillary barrier dysfunction. Furthermore, JAM-1 was markedly released into the plasma of polytrauma patients as early as 4 h after the trauma insult and significantly correlated with severity of disease and organ dysfunction (APACHE II and SOFA score). The data support an early injury- and time-dependent appearance of the barrier molecule JAM-1 in the circulation indicative of a commencing trauma-induced barrier dysfunction.
Collapse
|
22
|
Sun L, Liu S, Bao L, Li Y, Feng J, Liu Z. Claudin multigene family in channel catfish and their expression profiles in response to bacterial infection and hypoxia as revealed by meta-analysis of RNA-Seq datasets. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2015; 13:60-9. [PMID: 25681604 DOI: 10.1016/j.cbd.2015.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 01/14/2015] [Accepted: 01/14/2015] [Indexed: 11/19/2022]
Abstract
Claudins are one of the major groups of transmembrane proteins that play crucial roles in tight junctions. In addition to their function in the regulation of paracellular permeability, claudins are also involved in a number of biological processes related to pathogen infection, embryonic development, organ development and hypoxia response. Despite its importance, analyses of claudin genes in channel catfish have not been systematically performed. In this study, a total of 52 claudin genes were identified and characterized in channel catfish. Phylogenetic analyses were conducted to determine their identities and identify a number of lineage-specific claudin gene duplications in channel catfish. Expression profiles of catfish claudin genes in response to enteric septicemia of catfish (ESC) disease and hypoxia stress were determined by analyzing existing RNA-Seq datasets. Claudin genes were significantly down-regulated in the intestine at 3h post-infection, indicating that pathogens may disrupt the mucosal barrier by suppressing the expression of claudin genes. A total of six claudin genes were significantly regulated in the gill after hypoxia stress. Among them, the expressions of cldn-11b and cldn-10d were dramatically altered when comparing hypoxia tolerant fish with intolerant fish, though their specific roles involved in response to hypoxia stress remained unknown.
Collapse
Affiliation(s)
- Luyang Sun
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Shikai Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Lisui Bao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Yun Li
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Jianbin Feng
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, Program of Cell and Molecular Biosciences, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
23
|
Rodriguez Garcia M, Patel MV, Shen Z, Fahey JV, Biswas N, Mestecky J, Wira CR. Mucosal Immunity in the Human Female Reproductive Tract. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00108-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
24
|
Marshall LJ, Oguejiofor W, Willetts RS, Griffiths HR, Devitt A. Developing accurate models of the human airways. J Pharm Pharmacol 2014; 67:464-72. [DOI: 10.1111/jphp.12340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/19/2014] [Indexed: 12/14/2022]
Abstract
Abstract
Objectives
Particle delivery to the airways is an attractive prospect for many potential therapeutics, including vaccines. Developing strategies for inhalation of particles provides a targeted, controlled and non-invasive delivery route but, as with all novel therapeutics, in vitro and in vivo testing are needed prior to clinical use. Whilst advanced vaccine testing demands the use of animal models to address safety issues, the production of robust in vitro cellular models would take account of the ethical framework known as the 3Rs (Replacement, Reduction and Refinement of animal use), by permitting initial screening of potential candidates prior to animal use. There is thus a need for relevant, realistic in vitro models of the human airways.
Key findings
Our laboratory has designed and characterised a multi-cellular model of human airways that takes account of the conditions in the airways and recapitulates many salient features, including the epithelial barrier and mucus secretion.
Summary
Our human pulmonary models recreate many of the obstacles to successful pulmonary delivery of particles and therefore represent a valid test platform for screening compounds and delivery systems.
Collapse
Affiliation(s)
| | - Wilson Oguejiofor
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Rachel S Willetts
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Helen R Griffiths
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Andrew Devitt
- School of Life and Health Sciences, Aston University, Birmingham, UK
| |
Collapse
|
25
|
Bol L, Galas JC, Hillaireau H, Le Potier I, Nicolas V, Haghiri-Gosnet AM, Fattal E, Taverna M. A microdevice for parallelized pulmonary permeability studies. Biomed Microdevices 2014; 16:277-85. [PMID: 24337430 DOI: 10.1007/s10544-013-9831-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We describe a compartmentalized microdevice specifically designed to perform permeability studies across a model of lung barrier. Epithelial cell barriers were reproduced by culturing Calu-3 cells at the air-liquid interface (AIC) in 1 mm² microwells made from a perforated glass slide with an embedded porous membrane. We created a single basolateral reservoir for all microwells which eliminated the need to renew the growth medium during the culture growth phase. To perform drug permeability studies on confluent cell layers, the cell culture slide was aligned and joined to a collection platform consisting in 35 μL collection reservoirs connected at the top and bottom with microchannels. The integrity and functionality of the cell barriers were demonstrated by measurement of trans-epithelial electrical resistance (TEER), confocal imaging and permeability assays of ¹⁴C-sucrose. Micro-cell barriers were able to form confluent layers in 1 week, demonstrating a similar bioelectrical evolution as the Transwell systems used as controls. Tight junctions were observed throughout the cell-cell interfaces, and the low permeability coefficients of ¹⁴C-sucrose confirmed their functional presence, creating a primary barrier to the diffusion of solutes. This microdevice could facilitate the monitoring of biomolecule transport and the screening of formulations promoting their passage across the pulmonary barrier, in order to select candidates for pulmonary administration to patients.
Collapse
Affiliation(s)
- Ludivine Bol
- Faculté de Pharmacie, Université Paris-Sud, 92290, Châtenay-Malabry, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Snyder RJ, Hussain S, Rice AB, Garantziotis S. Multiwalled carbon nanotubes induce altered morphology and loss of barrier function in human bronchial epithelium at noncytotoxic doses. Int J Nanomedicine 2014; 9:4093-105. [PMID: 25187712 PMCID: PMC4149455 DOI: 10.2147/ijn.s65567] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Multiwalled carbon nanotubes (MWCNTs) have seen increasing application in consumer products over the past decade, resulting in an increasing risk of human exposure. While numerous toxicological studies have been performed using acute high doses of various carbonaceous nanomaterials, the effects of longer-term, low doses of MWCNTs remain relatively unexplored. This study examined bronchoscopy-derived healthy human bronchial epithelial cells exposed in submerged culture to noncytotoxic doses of MWCNTs over 7 days. Under these conditions, doses as low as 3 μg/mL caused altered cell morphology, superficially resembling fibroblasts. Electrical impedance of the epithelial monolayer was greatly reduced following MWCNT exposure. However, Western blot and polymerase chain reaction showed no elevated expression of the fibroblast markers, vimentin, α-smooth muscle actin, or fibronectin, indicating that a mechanism other than epithelial-mesenchymal transition may be responsible for the changes. Phalloidin and tubulin immunostaining showed disruption of the cytoskeleton, and confocal imaging showed a reduction of the tight junction proteins, zona occludens 1 and occludin. We propose that MWCNTs interfere with the cytoskeleton of the lung epithelium, which can result in a harmful reduction in barrier function over time, even at noncytotoxic doses.
Collapse
Affiliation(s)
- Ryan J Snyder
- Clinical Research Unit, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC, USA
| | - Salik Hussain
- Clinical Research Unit, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC, USA
| | - Annette B Rice
- Clinical Research Unit, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC, USA
| | - Stavros Garantziotis
- Clinical Research Unit, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, NC, USA
| |
Collapse
|
27
|
Wira CR, Fahey JV, Rodriguez-Garcia M, Shen Z, Patel MV. Regulation of mucosal immunity in the female reproductive tract: the role of sex hormones in immune protection against sexually transmitted pathogens. Am J Reprod Immunol 2014; 72:236-58. [PMID: 24734774 PMCID: PMC4351777 DOI: 10.1111/aji.12252] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/15/2014] [Indexed: 01/01/2023] Open
Abstract
The immune system in the female reproductive tract (FRT) does not mount an attack against human immunodeficiency virus (HIV) or other sexually transmitted infections (STI) with a single endogenously produced microbicide or with a single arm of the immune system. Instead, the body deploys dozens of innate antimicrobials to the secretions of the FRT. Working together, these antimicrobials along with mucosal antibodies attack viral, bacterial, and fungal targets. Within the FRT, the unique challenges of protection against sexually transmitted pathogens coupled with the need to sustain the development of an allogeneic fetus, has evolved in such a way that sex hormones precisely regulate immune function to accomplish both tasks. The studies presented in this review demonstrate that estradiol (E2 ) and progesterone secreted during the menstrual cycle act both directly and indirectly on epithelial cells, fibroblasts and immune cells in the reproductive tract to modify immune function in a way that is unique to specific sites throughout the FRT. As presented in this review, studies from our laboratory and others demonstrate that the innate and adaptive immune systems are under hormonal control, that protection varies with the stage of the menstrual cycle and as such, is dampened during the secretory stage of the cycle to optimize conditions for fertilization and pregnancy. In doing so, a window of STI vulnerability is created during which potential pathogens including HIV enter the reproductive tract to infect host targets.
Collapse
Affiliation(s)
- Charles R Wira
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, One Medical Center Drive, Lebanon, NH, USA
| | | | | | | | | |
Collapse
|
28
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg H, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX-ALTERNATIVES TO ANIMAL EXPERIMENTATION 2014. [PMID: 25027500 DOI: 10.14573/altex1406111] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
|
29
|
Dekali S, Gamez C, Kortulewski T, Blazy K, Rat P, Lacroix G. Assessment of an in vitro model of pulmonary barrier to study the translocation of nanoparticles. Toxicol Rep 2014; 1:157-171. [PMID: 28962236 PMCID: PMC5598380 DOI: 10.1016/j.toxrep.2014.03.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/10/2014] [Accepted: 03/10/2014] [Indexed: 12/22/2022] Open
Abstract
As the lung is one of the main routes of exposure to manufactured nanoparticles, we developed an in vitro model resembling the alveolo-capillary barrier for the study of nanoparticle translocation. In order to provide a relevant and ethical in vitro model, cost effective and easy-to-implement human cell lines were used. Pulmonary epithelial cells (Calu-3 cell line) and macrophages (THP-1 differentiated cells) were cultivated on the apical side and pulmonary endothelial cells (HPMEC-ST1.6R cell line) on the basal side of a microporous polyester membrane (Transwell®). Translocation of non-functionalized (51 and 110 nm) and aminated (52 nm) fluorescent polystyrene (PS) nanobeads was studied in this system. The use of Calu-3 cells allowed high transepithelial electrical resistance (TEER) values (>1000 Ω cm2) in co-cultures with or without macrophages. After 24 h of exposure to non-cytotoxic concentrations of non-functionalized PS nanobeads, the relative TEER values (%/t0) were significantly decreased in co-cultures. Epithelial cells and macrophages were able to internalize PS nanobeads. Regarding translocation, Transwell® membranes per se limit the passage of nanoparticles between apical and basal side. However, small non-functionalized PS nanobeads (51 nm) were able to translocate as they were detected in the basal side of co-cultures. Altogether, these results show that this co-culture model present good barrier properties allowing the study of nanoparticle translocation but research effort need to be done to improve the neutrality of the porous membrane delimitating apical and basal sides of the model.
Collapse
Affiliation(s)
- Samir Dekali
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes (PRES Sorbonne Paris Cité), 75270 Paris Cedex 06, France
| | - Christelle Gamez
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
| | - Thierry Kortulewski
- CEA, DSV, iRCM, Plateforme imagerie photonique, 92260 Fontenay-aux-Roses, France
| | - Kelly Blazy
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
| | - Patrice Rat
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes (PRES Sorbonne Paris Cité), 75270 Paris Cedex 06, France
| | - Ghislaine Lacroix
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
- Corresponding author at: INERIS, Parc technologique ALATA, BP2, 60550, Verneuil-en-Halatte, France. Tel.: +33 3 44 55 63 15; fax: +33 3 44 55 66 05
| |
Collapse
|
30
|
Alépée N, Bahinski A, Daneshian M, De Wever B, Fritsche E, Goldberg A, Hansmann J, Hartung T, Haycock J, Hogberg HT, Hoelting L, Kelm JM, Kadereit S, McVey E, Landsiedel R, Leist M, Lübberstedt M, Noor F, Pellevoisin C, Petersohn D, Pfannenbecker U, Reisinger K, Ramirez T, Rothen-Rutishauser B, Schäfer-Korting M, Zeilinger K, Zurich MG. State-of-the-art of 3D cultures (organs-on-a-chip) in safety testing and pathophysiology. ALTEX 2014; 31:441-77. [PMID: 25027500 PMCID: PMC4783151 DOI: 10.14573/altex.1406111] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/30/2014] [Indexed: 02/02/2023]
Abstract
Integrated approaches using different in vitro methods in combination with bioinformatics can (i) increase the success rate and speed of drug development; (ii) improve the accuracy of toxicological risk assessment; and (iii) increase our understanding of disease. Three-dimensional (3D) cell culture models are important building blocks of this strategy which has emerged during the last years. The majority of these models are organotypic, i.e., they aim to reproduce major functions of an organ or organ system. This implies in many cases that more than one cell type forms the 3D structure, and often matrix elements play an important role. This review summarizes the state of the art concerning commonalities of the different models. For instance, the theory of mass transport/metabolite exchange in 3D systems and the special analytical requirements for test endpoints in organotypic cultures are discussed in detail. In the next part, 3D model systems for selected organs--liver, lung, skin, brain--are presented and characterized in dedicated chapters. Also, 3D approaches to the modeling of tumors are presented and discussed. All chapters give a historical background, illustrate the large variety of approaches, and highlight up- and downsides as well as specific requirements. Moreover, they refer to the application in disease modeling, drug discovery and safety assessment. Finally, consensus recommendations indicate a roadmap for the successful implementation of 3D models in routine screening. It is expected that the use of such models will accelerate progress by reducing error rates and wrong predictions from compound testing.
Collapse
Affiliation(s)
| | - Anthony Bahinski
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA
| | - Mardas Daneshian
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany
| | | | - Ellen Fritsche
- Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Alan Goldberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - John Haycock
- Department of Materials Science of Engineering, University of Sheffield, Sheffield, UK
| | - Helena T. Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, USA
| | - Lisa Hoelting
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | | | - Suzanne Kadereit
- Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Emily McVey
- Board for the Authorization of Plant Protection Products and Biocides, Wageningen, The Netherlands
| | | | - Marcel Leist
- Center for Alternatives to Animal Testing – Europe (CAAT-Europe), University of Konstanz, Konstanz, Germany,Doerenkamp-Zbinden Chair of in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marc Lübberstedt
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Fozia Noor
- Biochemical Engineering, Saarland University, Saarbruecken, Germany
| | | | | | | | | | - Tzutzuy Ramirez
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Monika Schäfer-Korting
- Institute for Pharmacy (Pharmacology and Toxicology), Freie Universität Berlin, Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Marie-Gabriele Zurich
- Department of Physiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland,Swiss Center for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
31
|
Gehr P, Clift MJD, Brandenberger C, Lehmann A, Herzog F, Rothen-Rutishauser B. Endocytosis of environmental and engineered micro- and nanosized particles. Compr Physiol 2013; 1:1159-74. [PMID: 23733639 DOI: 10.1002/cphy.c100035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There are many studies with cells to find out how particles interact with them. In contrast to micronsized particles, which are actively taken up by phagocytosis or macropinocytosis, nanosized particles may be taken up by cells through different endocytic pathways or by another, yet to be defined mechanism. There is increasing evidence that it is the nanosized particles, which are a particular risk because of their high content of organic chemicals and their pro-oxidative potential due to the high surface-to-volume ratio of the particles as compared to the bulk material. It is the goal of this article to create an understanding for the interaction of particles with biological systems, with particular consideration of the interaction of nanoparticles (NPs) with lung cells. One is attempting to understand, how NPs interact with cellular membranes, as it is hardly known, how they are taken up by cells, how they are trafficking in cells, and how they interact with subcellular compartments, such as with mitochondria or with the nucleus. Cells tend to defend themselves against any foreign material, which is taken up. In general, they try to eliminate particulate intruders and this is what they usually manage with micronsized particles. However, with NPs it is different. NPs may not be eliminated easily, and, hence may stimulate the cells to react in an unfavorable way. What we can learn is that NPs behave differently than microparticles.
Collapse
Affiliation(s)
- Peter Gehr
- Institute of Anatomy, University of Bern, Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
32
|
Bersani I, Kunzmann S, Speer CP. Immunomodulatory properties of surfactant preparations. Expert Rev Anti Infect Ther 2013; 11:99-110. [PMID: 23428105 DOI: 10.1586/eri.12.156] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Surfactant replacement significantly decreased acute pulmonary morbidity and mortality among preterm neonates with respiratory distress syndrome. Besides improving lung function and oxygenation, surfactant is also a key modulator of pulmonary innate and acquired immunity regulating lung inflammatory processes. In this review, we describe the immunomodulatory features of surfactant preparations. Various surfactant preparations decrease the proinflammatory cytokine and chemokine release, the oxidative burst activity, and the nitric oxide production in lung inflammatory cells such as alveolar neutrophils, monocytes and macrophages; they also affect lymphocyte proliferative response and immunoglobulin production, as well as natural killer and lymphokine-activated killer cell activity. In addition, surfactant preparations are involved in airway remodeling, as they decrease lung fibroblast proliferation capacity and the release of mediators involved in remodeling. Moreover, they increase cell transepithelial resistance and VEGF synthesis in lung epithelial cells. A number of different signaling pathways and molecules are involved in these processes. Because the inhibition of local immune response may decrease lung injury, surfactant therapeutic efficacy may be related not only to its biophysical characteristics but, at least in part, to its anti-inflammatory features and its effects on remodeling processes. However, further studies are required to identify which surfactant preparation ensures the highest anti-inflammatory activity, thereby potentially decreasing the inflammatory process underlying respiratory distress syndrome. In perspective, detailed characterization of these anti-inflammatory effects could help to improve the next generation of surfactant preparations.
Collapse
Affiliation(s)
- Iliana Bersani
- University Children's Hospital, University of Würzburg, Germany
| | | | | |
Collapse
|
33
|
The degradation of airway tight junction protein under acidic conditions is probably mediated by transient receptor potential vanilloid 1 receptor. Biosci Rep 2013; 33:BSR20130087. [PMID: 24073800 PMCID: PMC3814059 DOI: 10.1042/bsr20130087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acidic airway microenvironment is one of the representative pathophysiological features of chronic inflammatory respiratory diseases. Epithelial barrier function is maintained by TJs (tight junctions), which act as the first physical barrier against the inhaled substances and pathogens of airway. As previous studies described, acid stress caused impaired epithelial barriers and led the hyperpermeability of epithelium. However, the specific mechanism is still unclear. We have showed previously the existence of TRPV (transient receptor potential vanilloid) 1 channel in airway epithelium, as well as its activation by acidic stress in 16HBE cells. In this study, we explored the acidic stress on airway barrier function and TJ proteins in vitro with 16HBE cell lines. Airway epithelial barrier function was determined by measuring by TER (trans-epithelial electrical resistance). TJ-related protein [claudin-1, claudin-3, claudin-4, claudin-5, claudin-7 and ZO-1 (zonula occluden 1)] expression was examined by western blotting of insoluble fractions of cell extraction. The localization of TJ proteins were visualized by immunofluorescent staining. Interestingly, stimulation by pH 6.0 for 8 h slightly increased the epithelial resistance in 16HBE cells insignificantly. However, higher concentration of hydrochloric acid (lower than pH 5.0) did reduce the airway epithelial TER of 16HBE cells. The decline of epithelial barrier function induced by acidic stress exhibited a TRPV1-[Ca2+]i-dependent pathway. Of the TJ proteins, claudin-3 and claudin-4 seemed to be sensitive to acidic stress. The degradation of claudin-3 and claudin-4 induced by acidic stress could be attenuated by the specific TRPV1 blocker or intracellular Ca2+ chelator BAPTA/AM [1,2-bis-(o-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis(acetoxymethyl ester)].
Collapse
|
34
|
Klein SG, Serchi T, Hoffmann L, Blömeke B, Gutleb AC. An improved 3D tetraculture system mimicking the cellular organisation at the alveolar barrier to study the potential toxic effects of particles on the lung. Part Fibre Toxicol 2013; 10:31. [PMID: 23890538 PMCID: PMC3733942 DOI: 10.1186/1743-8977-10-31] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/21/2013] [Indexed: 12/16/2022] Open
Abstract
Background Exposure to fine and ultra-fine ambient particles is still a problem of concern in many industrialised parts of the world and the intensified use of nanotechnology may further increase exposure to small particles. Complex in vitro coculture systems may be valuable tools to study particle-induced processes and to extrapolate effects of particles on the lung. A system consisting of four different human cell lines which mimics the cell response of the alveolar surface in vitro was developed to study native aerosol exposure (Vitrocell™ chamber). The system is composed of an alveolar type-II cell line (A549), differentiated macrophage-like cells (THP-1), mast cells (HMC-1) and endothelial cells (EA.hy 926), seeded in a 3D-orientation on a microporous membrane. Results The spatial distribution of the cells in the tetraculture was analysed by confocal laser scanning microscopy (CLSM), showing a confluent layer of endothelial and epithelial cells on both sides of the transwell. Macrophage-like cells and mast cells can be found on top of the epithelial cells. The cells formed colonies under submerged conditions, which disappeared at the ALI. To evaluate the response to oxidative stress, the dichlorodihydrofluorescein diacetate (DCFH-DA) assay was used together with 2,2’-azobis-2-methyl-propanimidamide-dihydrochloride (AAPH) as inducer of oxidative stress. The tetraculture showed less induction of reactive oxygen species (ROS) production after being treated with a positive control compared to the monocultures of EA.hy 926, THP-1 and HMC-1. Submerged cultures showed elevated ROS and IL-8 levels compared to ALI cultures. The Vitrocell™ aerosol exposure system was not significantly influencing the viability. Using this system, cells were exposed to an aerosol of 50 nm SiO2-Rhodamine NPs in PBS. The distribution of the NPs in the tetraculture after exposure was evaluated by CLSM. Fluorescence from internalized particles was detected in CD11b-positive THP-1 cells only. Conclusion The system can be used in conjunction with a native aerosol exposure system and may finally lead to a more realistic judgement regarding the hazard of new compounds and/or new nano-scaled materials in the future. The results for the ROS production and IL-8 secretion suggest that submerged exposure may lead to an overestimation of observed effects.
Collapse
|
35
|
Hsia CCW, Schmitz A, Lambertz M, Perry SF, Maina JN. Evolution of air breathing: oxygen homeostasis and the transitions from water to land and sky. Compr Physiol 2013; 3:849-915. [PMID: 23720333 PMCID: PMC3926130 DOI: 10.1002/cphy.c120003] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Life originated in anoxia, but many organisms came to depend upon oxygen for survival, independently evolving diverse respiratory systems for acquiring oxygen from the environment. Ambient oxygen tension (PO2) fluctuated through the ages in correlation with biodiversity and body size, enabling organisms to migrate from water to land and air and sometimes in the opposite direction. Habitat expansion compels the use of different gas exchangers, for example, skin, gills, tracheae, lungs, and their intermediate stages, that may coexist within the same species; coexistence may be temporally disjunct (e.g., larval gills vs. adult lungs) or simultaneous (e.g., skin, gills, and lungs in some salamanders). Disparate systems exhibit similar directions of adaptation: toward larger diffusion interfaces, thinner barriers, finer dynamic regulation, and reduced cost of breathing. Efficient respiratory gas exchange, coupled to downstream convective and diffusive resistances, comprise the "oxygen cascade"-step-down of PO2 that balances supply against toxicity. Here, we review the origin of oxygen homeostasis, a primal selection factor for all respiratory systems, which in turn function as gatekeepers of the cascade. Within an organism's lifespan, the respiratory apparatus adapts in various ways to upregulate oxygen uptake in hypoxia and restrict uptake in hyperoxia. In an evolutionary context, certain species also become adapted to environmental conditions or habitual organismic demands. We, therefore, survey the comparative anatomy and physiology of respiratory systems from invertebrates to vertebrates, water to air breathers, and terrestrial to aerial inhabitants. Through the evolutionary directions and variety of gas exchangers, their shared features and individual compromises may be appreciated.
Collapse
Affiliation(s)
- Connie C W Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Rothen-Rutishauser B, Clift MJ, Jud C, Fink A, Wick P. Human epithelial cells in vitro – Are they an advantageous tool to help understand the nanomaterial-biological barrier interaction? ACTA ACUST UNITED AC 2012. [DOI: 10.1515/entl-2015-0004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstratThe human body can be exposed to nanomaterials through a variety of different routes. As nanomaterials get in contact with the skin, the gastrointestinal tract, and the respiratory tract, these biological compartments are acting as barriers to the passage of nano-sized materials into the organism. These structural and functional barriers are provided by the epithelia serving as an interface between biological compartments. In order to initiate the reduction, refinement and replacement of time consuming, expensive and stressful (to the animals) in vivo experimental approaches, many in vitro epithelial cell culture models have been developed during the last decades. This review therefore, focuses on the functional as well as structural aspects of epithelial cells as well as the most commonly used in vitro epithelial models of the primary biological barriers with which nanomaterials might come in contact with either occupationally, or during their manufacturing and application. The advantages and disadvantages of the different in vitro models are discussed in order to provide a clear overview as to whether or not epithelial cell cultures are an advantageous model to be used for basic mechanism and nanotoxicology research.
Collapse
|
38
|
Neuhaus W, Samwer F, Kunzmann S, Muellenbach RM, Wirth M, Speer CP, Roewer N, Förster CY. Lung endothelial cells strengthen, but brain endothelial cells weaken barrier properties of a human alveolar epithelium cell culture model. Differentiation 2012; 84:294-304. [PMID: 23023065 DOI: 10.1016/j.diff.2012.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 08/21/2012] [Accepted: 08/24/2012] [Indexed: 01/11/2023]
Abstract
The blood-air barrier in the lung consists of the alveolar epithelium, the underlying capillary endothelium, their basement membranes and the interstitial space between the cell layers. Little is known about the interactions between the alveolar and the blood compartment. The aim of the present study was to gain first insights into the possible interplay between these two neighbored cell layers. We established an in vitro Transwell model of the alveolar epithelium based on human cell line H441 and investigated the influence of conditioned medium obtained from human lung endothelial cell line HPMEC-ST1.6R on the barrier properties of the H441 layers. As control for tissue specificity H441 layers were exposed to conditioned medium from human brain endothelial cell line hCMEC/D3. Addition of dexamethasone was necessary to obtain stable H441 cell layers. Moreover, dexamethasone increased expression of cell type I markers (caveolin-1, RAGE) and cell type II marker SP-B, whereas decreased the transepithelial electrical resistance (TEER) in a concentration dependent manner. Soluble factors obtained from the lung endothelial cell line increased the barrier significantly proven by TEER values and fluorescein permeability on the functional level and by the differential expression of tight junctional proteins on the molecular level. In contrast to this, soluble factors derived from brain endothelial cells weakened the barrier significantly. In conclusion, soluble factors from lung endothelial cells can strengthen the alveolar epithelium barrier in vitro, which suggests communication between endothelial and epithelial cells regulating the integrity of the blood-air barrier.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Department of Anaesthesia and Critical Care, University Hospital Wuerzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Zheng J, Liu W, Fan Y, Ye X, Xia W, Wang H, Shi J, Xu G, Li H. Suppression of connexin 26 is related to protease-activated receptor 2-mediated pathway in patients with allergic rhinitis. Am J Rhinol Allergy 2012; 26:e5-9. [PMID: 22391066 DOI: 10.2500/ajra.2012.26.3740] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Connexin (Cx) 26 plays a key role in maintaining the integrity of tight junctions. However, the expression and modulation of Cx26 in allergic rhinitis (AR) has not been well understood. METHODS We detected the expression of Cx26 in house-dust mite (HDM)-sensitized AR patients and investigated the Cx26 production and modulation in primary human nasal epithelial cells (HNECs) and BEAS-2B cells after treatment with the allergen Der p 1 from Dermatophagoides pteronyssinus. RESULTS We found that the mRNA and protein levels of Cx26 were significantly down-regulated in AR patients compared with the control. Der p 1 was found to induce protease-activated receptor 2 (PAR2) expression and suppress Cx26 production significantly in vitro. PAR2 siRNA was shown to prevent the suppression of Cx26 induced by Der p 1 in BEAS-2B cells. CONCLUSION The suppression of Cx26 in HDM-sensitized AR patients is related to a PAR2-mediated pathway and might serve during the initiation and maintenance of AR. Targeting the PAR2-mediated Cx26 suppression may be a potential means of preventing allergic sensitization.
Collapse
Affiliation(s)
- Jing Zheng
- Allergy and Cancer Center, Otorhinolaryngology Hospital, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Müller L, Jaspers I. Epithelial cells, the "switchboard" of respiratory immune defense responses: effects of air pollutants. Swiss Med Wkly 2012; 142:w13653. [PMID: 22851042 DOI: 10.4414/smw.2012.13653] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
"Epimmunome", a term introduced recently by Swamy and colleagues, describes all molecules and pathways used by epithelial cells (ECs) to instruct immune cells. Today, we know that ECs are among the first sites within the human body to be exposed to pathogens (such as influenza viruses) and that the release of chemokine and cytokines by ECs is influenced by inhaled agents. The role of the ECs as a switchboard to initiate and regulate immune responses is altered through air pollutant exposure, such as ozone, tobacco smoke and diesel exhaust emissions. The details of the interplay between ECs and immune cells are not yet fully understood and need to be investigated further. Co-culture models, cell specific genetically-modified mice and the analysis of human biopsies provide great tools to gain knowledge about potential mechanisms. Increasing our understanding about the role of ECs in respiratory immunity may yield novel therapeutic targets to modulate downstream diseases.
Collapse
Affiliation(s)
- Loretta Müller
- Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7310, USA.
| | | |
Collapse
|
41
|
Simet SM, Wyatt TA, DeVasure J, Yanov D, Allen-Gipson D, Sisson JH. Alcohol increases the permeability of airway epithelial tight junctions in Beas-2B and NHBE cells. Alcohol Clin Exp Res 2011; 36:432-42. [PMID: 21950588 DOI: 10.1111/j.1530-0277.2011.01640.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Tight junctions form a continuous belt-like structure between cells and act to regulate paracellular signaling. Protein kinase C (PKC) has been shown to regulate tight junction assembly and disassembly and is activated by alcohol. Previous research has shown that alcohol increases the permeability of tight junctions in lung alveolar cells. However, little is known about alcohol's effect on tight junctions in epithelium of the conducting airways. We hypothesized that long-term alcohol exposure reduces zonula occluden-1 (ZO-1) and claudin-1 localization at the cell membrane and increases permeability through a PKC-dependent mechanism. METHODS To test this hypothesis, we exposed normal human bronchial epithelial (NHBE) cells, cells from a human bronchial epithelial transformed cell line (Beas-2B), and Beas-2B expressing a PKCα dominant negative (DN) to alcohol (20, 50, and 100 mM) for up to 48 hours. Immunofluorescence was used to assess changes in ZO-1, claudin-1, claudin-5, and claudin-7 localization. Electric cell-substrate impedance sensing was used to measure the permeability of tight junctions between monolayers of NHBE, Beas-2B, and DN cells. RESULTS Alcohol increased tight junction permeability in a concentration-dependent manner and decreased ZO-1, claudin-1, claudin-5, and claudin-7 localization at the cell membrane. To determine a possible signaling mechanism, we measured the activity of PKC isoforms (alpha, delta, epsilon, and zeta). PKCα activity significantly increased in Beas-2B cells from 1 to 6 hours of 100 mM alcohol exposure, while PKCζ activity significantly decreased at 1 hour and increased at 3 hours. Inhibiting PKCα with Gö-6976 prevented the alcohol-induced protein changes in both ZO-1 and claudin-1 at the cell membrane. PKCα DN Beas-2B cells were resistant to alcohol-induced protein alterations. CONCLUSIONS These results suggest that alcohol disrupts ZO-1, claudin-1, claudin-5, and claudin-7 through the activation of PKCα, leading to an alcohol-induced "leakiness" in bronchial epithelial cells. Such alcohol-induced airway-leak state likely contributes to the impaired airway host defenses associated with acute and chronic alcohol ingestion.
Collapse
Affiliation(s)
- Samantha M Simet
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center, Omaha, Nebraska 68198-5910, USA
| | | | | | | | | | | |
Collapse
|
42
|
Klein SG, Hennen J, Serchi T, Blömeke B, Gutleb AC. Potential of coculture in vitro models to study inflammatory and sensitizing effects of particles on the lung. Toxicol In Vitro 2011; 25:1516-34. [PMID: 21963807 DOI: 10.1016/j.tiv.2011.09.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2011] [Revised: 07/18/2011] [Accepted: 09/06/2011] [Indexed: 12/30/2022]
Abstract
Exposure to particulate matter (PM) like nanoparticles (NPs) has increased in the last century due to increased combustion processes, road traffic, etc. In addition, the progress in chemical and cosmetic industry led to many new compounds, e.g. fragrances, which humans are exposed to every day. Many chemicals are known to act as contact and some as respiratory sensitizers, causing allergic reactions. Exposure to small particles of less than 100 nm in diameter is linked with an increased risk of respiratory diseases, such as asthma or rhinitis. To date already more than 1000 customer products contain eNPs without knowing much about the health effects. In comparison to chemicals, the mechanisms by which PM and eNPs can cause sensitization are still not fully understood. Validated and regulatory accepted in vitro models to assess this hazard in its full range are still missing. While a huge number of animal studies contributed to our knowledge about sensitization processes, knowledge on involved cellular mechanisms is still limited. In this review relevant in vitro models to study and elucidate these mechanisms in more detail are presented and their potential to serve as part of a tiered testing strategy is discussed.
Collapse
Affiliation(s)
- Sebastian G Klein
- Department Environment and Agro-biotechnologies (EVA), Centre de Recherche Public, Gabriel Lippmann, 41 rue du Brill, L-4422 Belvaux, Luxembourg
| | | | | | | | | |
Collapse
|
43
|
Carter CJ. Pathogen and autoantigen homologous regions within the cystic fibrosis transmembrane conductance regulator (CFTR) protein suggest an autoimmune treatable component of cystic fibrosis. ACTA ACUST UNITED AC 2011; 62:197-214. [DOI: 10.1111/j.1574-695x.2011.00803.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
44
|
Sun YH, Reid B, Fontaine JH, Miller LA, Hyde DM, Mogilner A, Zhao M. Airway epithelial wounds in rhesus monkey generate ionic currents that guide cell migration to promote healing. J Appl Physiol (1985) 2011; 111:1031-41. [PMID: 21719726 DOI: 10.1152/japplphysiol.00915.2010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Damage to the respiratory epithelium is one of the most critical steps to many life-threatening diseases, such as acute respiratory distress syndrome and chronic obstructive pulmonary disease. The mechanisms underlying repair of the damaged epithelium have not yet been fully elucidated. Here we provide experimental evidence suggesting a novel mechanism for wound repair: endogenous electric currents. It is known that the airway epithelium maintains a voltage difference referred to as the transepithelial potential. Using a noninvasive vibrating probe, we demonstrate that wounds in the epithelium of trachea from rhesus monkeys generate significant outward electric currents. A small slit wound produced an outward current (1.59 μA/cm(2)), which could be enhanced (nearly doubled) by the ion transport stimulator aminophylline. In addition, inhibiting cystic fibrosis transmembrane conductance regulator (CFTR) with CFTR(Inh)-172 significantly reduced wound currents (0.17 μA/cm(2)), implicating an important role of ion transporters in wound induced electric potentials. Time-lapse video microscopy showed that applied electric fields (EFs) induced robust directional migration of primary tracheobronchial epithelial cells from rhesus monkeys, towards the cathode, with a threshold of <23 mV/mm. Reversal of the field polarity induced cell migration towards the new cathode. We further demonstrate that application of an EF promoted wound healing in a monolayer wound healing assay. Our results suggest that endogenous electric currents at sites of tracheal epithelial injury may direct cell migration, which could benefit restitution of damaged airway mucosa. Manipulation of ion transport may lead to novel therapeutic approaches to repair damaged respiratory epithelium.
Collapse
Affiliation(s)
- Yao-Hui Sun
- Department of Dermatology, School of Medicine, Univ. of California at Davis CA, Sacramento, CA 95817, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Hyun SW, Anglin IE, Liu A, Yang S, Sorkin JD, Lillehoj E, Tonks NK, Passaniti A, Goldblum SE. Diverse injurious stimuli reduce protein tyrosine phosphatase-μ expression and enhance epidermal growth factor receptor signaling in human airway epithelia. Exp Lung Res 2011; 37:327-43. [PMID: 21649524 DOI: 10.3109/01902148.2011.566673] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In response to injury, airway epithelia utilize an epidermal growth factor (EGF) receptor (EGFR) signaling program to institute repair and restitution. Protein tyrosine phosphatases (PTPs) counterregulate EGFR autophosphorylation and downstream signaling. PTPμ is highly expressed in lung epithelia and can be localized to intercellular junctions where its ectodomain homophilically interacts with PTPμ ectodomain expressed on neighboring cells. We asked whether PTPμ expression might be altered in response to epithelial injury and whether altered PTPμ expression might influence EGFR signaling. In A549 cells, diverse injurious stimuli dramatically reduced PTPμ protein expression. Under basal conditions, small interfering RNA (siRNA)-induced silencing of PTPμ increased EGFR Y992 and Y1068 phosphorylation. In the presence of EGF, PTPμ knockdown increased EGFR Y845, Y992, Y1045, Y1068, Y1086, and Y1173 but not Y1148 phosphorylation. Reduced PTPμ expression increased EGF-stimulated phosphorylation of Y992, a docking site for phospholipase C (PLC)γ(1), activation of PLCγ(1) itself, and increased cell migration in both wounding and chemotaxis assays. In contrast, overexpression of PTPμ decreased EGF-stimulated EGFR Y992 and Y1068 phosphorylation. Therefore, airway epithelial injury profoundly reduces PTPμ expression, and PTPμ depletion selectively increases phosphorylation of specific EGFR tyrosine residues, PLCγ(1) activation, and cell migration, providing a novel mechanism through which epithelial integrity may be restored.
Collapse
Affiliation(s)
- Sang W Hyun
- Department of Medicine, Mucosal Biology Research Center, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Tight junctions are the most apically localized part of the epithelial junctional complex. They regulate the permeability and polarity of cell layers and create compartments in cell membranes. Claudins are structural molecules of tight junctions. There are 27 claudins known, and expression of different claudins is responsible for changes in the electrolyte and solute permeability in cells layers. Studies have shown that claudins and tight junctions also protect multicellular organisms from infections and that some infectious agents may use claudins as targets to invade and weaken the host's defense. In neoplastic diseases, claudin expression may be up- or downregulated. Since their expression is associated with specific tumor types or with specific locations of tumors to a certain degree, they can, in a restricted sense, also be used as tumor markers. However, the regulation of claudin expression is complex involving growth factors and integrins, protein kinases, proto-oncogens and transcription factors. In this review, the significance of claudins is discussed in lung disease and development.
Collapse
Affiliation(s)
- Ylermi Soini
- Department of Pathology and Forensic Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, School of Medicine, University of Eastern Finland, Cancer Center of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
47
|
Simões T, Charro N, Blonder J, Faria D, Couto FM, Chan KC, Waybright T, Isaaq HJ, Veenstra TD, Penque D. Molecular profiling of the human nasal epithelium: A proteomics approach. J Proteomics 2011; 75:56-69. [PMID: 21621024 PMCID: PMC7185466 DOI: 10.1016/j.jprot.2011.05.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 05/05/2011] [Indexed: 12/25/2022]
Abstract
A comprehensive proteomic profiling of nasal epithelium (NE) is described. This study relies on simple subcellular fractionation used to obtain soluble- and membrane-enriched fractions followed by 2-dimensional liquid chromatography (2D-LC) separation and tandem mass spectrometry (MS/MS). The cells were collected using a brushing technique applied on NE of clinically evaluated volunteers. Subsequently, the soluble- and the membrane-protein enriched fractions were prepared and analyzed in parallel using 2D-LC-MS/MS. In a set of 1482 identified proteins, 947 (63.9%) proteins were found to be associated to membrane fraction. Grand average hydropathy value index (GRAVY) analysis, the transmembrane protein mapping and annotations of primary location deposited in the Human Protein Reference Database (HPRD) confirmed an enrichment of hydrophobic proteins on this dataset. Ingenuity Pathway Analysis (IPA) of soluble fraction revealed an enrichment of molecular and cellular functions associated with cell death, protein folding and drug metabolism while in membrane fraction showed an enrichment of functions associated with molecular transport, protein trafficking and cell-to-cell signaling and interaction. The IPA showed similar enrichment of functions associated with cellular growth and proliferation in both soluble and membrane subproteomes. This finding was in agreement with protein content analysis using exponentially modified protein abundance index (emPAI). A comparison of our data with previously published studies focusing on respiratory tract epithelium revealed similarities related to identification of proteins associated with physical barrier function and immunological defence. In summary, we extended the NE molecular profile by identifying and characterizing proteins associated to pivotal functions of a respiratory epithelium, including the control of fluid volume and ionic composition at the airways' surface, physical barrier maintenance, detoxification and immunological defence. The extent of similarities supports the applicability of a less invasive analysis of NE to assess prognosis and treatment response of lung diseases such as asthma, cystic fibrosis and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Tânia Simões
- Laboratório de Proteómica, Departamento de Genética, Instituto Nacional de Saúde Dr. Ricardo Jorge, INSA I.P., Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Blank F, Wehrli M, Lehmann A, Baum O, Gehr P, von Garnier C, Rothen-Rutishauser BM. Macrophages and dendritic cells express tight junction proteins and exchange particles in an in vitro model of the human airway wall. Immunobiology 2011; 216:86-95. [DOI: 10.1016/j.imbio.2010.02.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 02/24/2010] [Accepted: 02/24/2010] [Indexed: 10/19/2022]
|
49
|
Swain RJ, Kemp SJ, Goldstraw P, Tetley TD, Stevens MM. Assessment of cell line models of primary human cells by Raman spectral phenotyping. Biophys J 2010; 98:1703-11. [PMID: 20409492 DOI: 10.1016/j.bpj.2009.12.4289] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 11/27/2009] [Accepted: 12/09/2009] [Indexed: 12/19/2022] Open
Abstract
Researchers have previously questioned the suitability of cell lines as models for primary cells. In this study, we used Raman microspectroscopy to characterize live A549 cells from a unique molecular biochemical perspective to shed light on their suitability as a model for primary human pulmonary alveolar type II (ATII) cells. We also investigated a recently developed transduced type I (TT1) cell line as a model for alveolar type I (ATI) cells. Single-cell Raman spectra provide unique biomolecular fingerprints that can be used to characterize cellular phenotypes. A multivariate statistical analysis of Raman spectra indicated that the spectra of A549 and TT1 cells are characterized by significantly lower phospholipid content compared to ATII and ATI spectra because their cytoplasm contains fewer surfactant lamellar bodies. Furthermore, we found that A549 spectra are statistically more similar to ATI spectra than to ATII spectra. The spectral variation permitted phenotypic classification of cells based on Raman spectral signatures with >99% accuracy. These results suggest that A549 cells are not a good model for ATII cells, but TT1 cells do provide a reasonable model for ATI cells. The findings have far-reaching implications for the assessment of cell lines as suitable primary cellular models in live cultures.
Collapse
Affiliation(s)
- Robin J Swain
- Department of Materials, Imperial College London, London, United Kingdom
| | | | | | | | | |
Collapse
|
50
|
Wira CR, Fahey JV, Ghosh M, Patel MV, Hickey DK, Ochiel DO. Sex hormone regulation of innate immunity in the female reproductive tract: the role of epithelial cells in balancing reproductive potential with protection against sexually transmitted pathogens. Am J Reprod Immunol 2010; 63:544-65. [PMID: 20367623 DOI: 10.1111/j.1600-0897.2010.00842.x] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The immune system in the female reproductive tract (FRT) does not mount an attack against HIV or other sexually transmitted infections (STI) with a single endogenously produced microbicide or with a single arm of the immune system. Instead, the body deploys dozens of innate antimicrobials to the secretions of the female reproductive tract. Working together, these antimicrobials along with mucosal antibodies attack many different viral, bacterial and fungal targets. Within the FRT, the unique challenges of protection against sexually transmitted pathogens coupled with the need to sustain the development of an allogeneic fetus have evolved in such a way that sex hormones precisely regulate immune function to accomplish both tasks. The studies presented in this review demonstrate that estradiol and progesterone secreted during the menstrual cycle act both directly and indirectly on epithelial cells and other immune cells in the reproductive tract to modify immune function in a way that is unique to specific sites throughout the FRT. As presented in this review, studies from our laboratory and others demonstrate that the innate immune response is under hormonal control, varies with the stage of the menstrual cycle, and as such is suppressed at mid-cycle to optimize conditions for successful fertilization and pregnancy. In doing so, a window of STI vulnerability is created during which potential pathogens including HIV enter the reproductive tract to infect host targets.
Collapse
Affiliation(s)
- Charles R Wira
- Department of Physiology, Dartmouth Medical School, One Medical Center Drive, Lebanon, NH 03756, USA.
| | | | | | | | | | | |
Collapse
|