1
|
Pino JS, Alvarado PN, Larrea AM, Rojas W, Gomez-Lopera N. Analysis of cytotoxicity and genotoxicity of diesel exhaust PM2.5 generated from diesel and dual natural gas-diesel engines. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104638. [PMID: 39765323 DOI: 10.1016/j.etap.2025.104638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/26/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Diesel exhaust particles (DEPs) are atmospheric pollutants associated with adverse health effects. In response to their impact, natural gas (NG) has emerged as a promising alternative fuel due to its cleaner combustion. Although the cytotoxicity and genotoxicity of DEPs from diesel or NG engines have been extensively studied, the impact of dual natural gas-diesel systems remains unexplored. This study evaluated the toxicity of DEPs (PM2.5) emitted by an engine in diesel mode and dual natural gas-diesel mode on cellular parameters such as viability, apoptosis, oxidative stress, and DNA damage. The results showed that diesel DEPs reduced cell viability by up to 31 %, compared to a 19.2 % reduction with dual-mode DEPs. Apoptosis induction was also higher with diesel DEPs, with a 7 % increase compared to the dual mode. While dual-mode DEPs increased the production of reactive oxygen species (ROS) without causing DNA damage, diesel DEPs generated high ROS levels and measurable DNA damage. These differences could be attributed to the physicochemical characteristics of each mode, as diesel DEPs contained higher concentrations of polycyclic aromatic hydrocarbons (PAHs). This study addresses a research gap by quantifying the health effects of emissions from dual-fuel engines and highlights the potential of these systems to reduce DEP-induced toxicity.
Collapse
Affiliation(s)
- Juan Sebastian Pino
- Facultad de Ciencias Exactas y Naturales. Grupo de Genética Molecular, Universidad de Antioquia, Medellín, Colombia
| | - Pedro N Alvarado
- Facultad de Ingenierías. Grupo de Investigación Materiales Avanzados y Energía, Instituto Tecnológico Metropolitano, Medellín, Colombia
| | - Ana Maria Larrea
- Facultad de Ciencias Exactas y Naturales. Grupo de Genética Molecular, Universidad de Antioquia, Medellín, Colombia
| | - Winston Rojas
- Facultad de Ciencias Exactas y Naturales. Grupo de Genética Molecular, Universidad de Antioquia, Medellín, Colombia
| | - Natalia Gomez-Lopera
- Facultad de Medicina. Grupo de Genética Médica, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
2
|
Li J, Song H, Zhang L, Li J, Yang Y, Cui X, Mahfuza A, Cao Y, Hu X, Li C, Zhao Q, Tian S. Interaction of diesel exhaust particulate matter with mucins in simulated saliva fluids: Bioaccessibility of heavy metals and potential health risks. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:135811. [PMID: 39298947 DOI: 10.1016/j.jhazmat.2024.135811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Air pollution is one of the major environmental risks threatening human health, diesel exhaust particulate matter (DEPM) is an important source of urban air pollution, and oral ingestion is the primary route of exposure to atmospheric particulate matter. This study examined the bioaccessibility of Cr, Fe, and Zn in DEPM within simulated saliva fluids through in vitro experiments, interactions between the particles and mucins, and the mechanisms underlying the oxidative damage they cause. The results indicated that the interaction between DEPM and mucins altered the dispersibility, surface charge, and wettability of the particles, leading to increased release of heavy metals. Protein adsorption experiments and characterizations revealed that the adsorption of mucin by the particles resulted in a complexation reaction between the metals in the DEPM and the mucins, accompanied by fluorescence quenching of the protein. In addition, free radical assays and correlation analyses revealed that environmentally persistent free radicals generated by DEPM induce the production of reactive oxygen species (O2·-, HOOH, and·OH), which damage the secondary structure of mucins and increase the risk of oral diseases. Our study is the first to reveal the interaction between DEPM and mucins in saliva, elucidating the mechanisms of DEPM-induced oxidative damage. This is significant for understanding the oral health risks posed by the ingestion of atmospheric particulate matter.
Collapse
Affiliation(s)
- Jiao Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Haorang Song
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Linfeng Zhang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Jie Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yanlin Yang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xiangfen Cui
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Anjum Mahfuza
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yan Cao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Xuewei Hu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Chen Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qun Zhao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| | - Senlin Tian
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
3
|
Ji J, Jansen K, Kessler V, Seisenbaeva G, Gerde P, Malmlöf M, Palmberg L, Upadhyay S. Cell line-based in vitro models of normal and chronic bronchitis-like airway mucosa to study the toxic potential of aerosolized palladium nanoparticles. Front Med (Lausanne) 2024; 11:1422792. [PMID: 39440037 PMCID: PMC11493715 DOI: 10.3389/fmed.2024.1422792] [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: 04/24/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Background Physiologically relevant cell line-based models of human airway mucosa are needed to assess nanoparticle-mediated pulmonary toxicity for any xenbiotics expsoure study. Palladium nanoparticles (Pd-NP) originating from catalytic converters in vehicles pose health risks. We aimed to develop in vitro airway models to assess the toxic potential of Pd-NP in normal (Non-CB) and chronic bronchitis-like (CB-like) mucosa models. Methods Bronchial mucosa models were developed using Epithelial cells (16HBE: apical side) co-cultured with fibroblast (basal side) at an air-liquid interface. Furthermore, both Non-CB and CB-like (IL-13 treatment) models with increased numbers of goblet cells were used. The models were exposed to 3 different doses of aerosolized Pd-NP (0.2, 0.3, and 6 μg/cm2) using XposeALI® and clean air as a control. After 24 h of incubation, the expression of inflammatory (IL6, CXCL8, TNFα, and NFKB), oxidative stress (HMOX1, SOD3, GPx, and GSTA1), and tissue injury/repair (MMP9/TIMP1) markers was assessed using qRT-PCR. The secretion of CXCL-8 and the expression of a tissue injury/repair marker (MMP-9) were measured via ELISA. Results Significantly (p < 0.05) increased expressions of CXCL8, IL6, and NFKB were observed at the highest dose of Pd-NP in CB-like models. However, in Non-CB mucosa models, a maximum effect on TNFα and NFKB expression was observed at a medium Pd-NP dose. In Non-CB mucosa models, SOD3 showed a clear dose-dependent response to Pd-NP exposure, while GSTA1 expression was significantly increased (p < 0.05) only at the lowest dose of Pd-NP. The secretion of CXCL-8 increased in a dose-dependent manner in the Non-CB mucosa models following exposure to Pd-NP. In CB-like models, exposure to high concentrations of Pd-NP significantly increased the release of MMP-9 compared to that in other exposure groups. Conclusion The combination of our Non-CB and CB-like mucosa models with the XposeALI® system for aerosolized nanoparticle exposure closely mimics in vivo lung environments and cell-particle interactions. Results from these models, utilizing accessible cell lines, will maximize the reliability of in vitro findings in human health risk assessment.
Collapse
Affiliation(s)
- Jie Ji
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Katja Jansen
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vadim Kessler
- Inorganic Bionanotechnology Unit, Department of Chemistry and Biotechnology, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Gulaim Seisenbaeva
- Inorganic Bionanotechnology Unit, Department of Chemistry and Biotechnology, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Per Gerde
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Inhalation Sciences Sweden AB, Stockholm, Sweden
| | | | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Lukyanchuk A, Muraki N, Kawai T, Sato T, Hata K, Ito T, Tajima A. Long-term exposure to diesel exhaust particles induces concordant changes in DNA methylation and transcriptome in human adenocarcinoma alveolar basal epithelial cells. Epigenetics Chromatin 2024; 17:24. [PMID: 39103936 DOI: 10.1186/s13072-024-00549-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Diesel exhaust particles (DEP), which contain hazardous compounds, are emitted during the combustion of diesel. As approximately one-third of the vehicles worldwide use diesel, there are growing concerns about the risks posed by DEP to human health. Long-term exposure to DEP is associated with airway hyperresponsiveness, pulmonary fibrosis, and inflammation; however, the molecular mechanisms behind the effects of DEP on the respiratory tract are poorly understood. Such mechanisms can be addressed by examining transcriptional and DNA methylation changes. Although several studies have focused on the effects of short-term DEP exposure on gene expression, research on the transcriptional effects and genome-wide DNA methylation changes caused by long-term DEP exposure is lacking. Hence, in this study, we investigated transcriptional and DNA methylation changes in human adenocarcinoma alveolar basal epithelial A549 cells caused by prolonged exposure to DEP and determined whether these changes are concordant. RESULTS DNA methylation analysis using the Illumina Infinium MethylationEPIC BeadChips showed that the methylation levels of DEP-affected CpG sites in A549 cells changed in a dose-dependent manner; the extent of change increased with increasing dose reaching the statistical significance only in samples exposed to 30 µg/ml DEP. Four-week exposure to 30 µg/ml of DEP significantly induced DNA hypomethylation at 24,464 CpG sites, which were significantly enriched for DNase hypersensitive sites, genomic regions marked by H3K4me1 and H3K27ac, and several transcription factor binding sites. In contrast, 9,436 CpG sites with increased DNA methylation levels were significantly overrepresented in genomic regions marked by H3K27me3 as well as H3K4me1 and H3K27ac. In parallel, gene expression profiling by RNA sequencing demonstrated that long-term exposure to DEP altered the expression levels of 2,410 genes, enriching 16 gene sets including Xenobiotic metabolism, Inflammatory response, and Senescence. In silico analysis revealed that the expression levels of 854 genes correlated with the methylation levels of the DEP-affected cis-CpG sites. CONCLUSIONS To our knowledge, this is the first report of genome-wide transcriptional and DNA methylation changes and their associations in A549 cells following long-term exposure to DEP.
Collapse
Affiliation(s)
- Alexandra Lukyanchuk
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Krasnoyarsk State Medical University Named After Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Naomi Muraki
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
- Department of Human Biology and Anatomy, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, Tokyo, Japan
- Department of Human Molecular Genetics, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tsuyoshi Ito
- Health Effects Research Group, Environment Research Division, Japan Automobile Research Institute, Tsukuba, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| |
Collapse
|
5
|
Cattani-Cavalieri I, Trombetta-Lima M, Yan H, Manzano-Covarrubias AL, Baarsma HA, Oun A, van der Veen MM, Oosterhout E, Dolga AM, Ostrom RS, Valenca SS, Schmidt M. Diesel exhaust particles alter mitochondrial bioenergetics and cAMP producing capacity in human bronchial epithelial cells. FRONTIERS IN TOXICOLOGY 2024; 6:1412864. [PMID: 39118833 PMCID: PMC11306203 DOI: 10.3389/ftox.2024.1412864] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Air pollution from diesel combustion is linked in part to the generation of diesel exhaust particles (DEP). DEP exposure induces various processes, including inflammation and oxidative stress, which ultimately contribute to a decline in lung function. Cyclic AMP (cAMP) signaling is critical for lung homeostasis. The impact of DEP on cAMP signaling is largely unknown. Methods: We exposed human bronchial epithelial (BEAS-2B) cells to DEP for 24-72 h and evaluated mitochondrial bioenergetics, markers of oxidative stress and inflammation and the components of cAMP signaling. Mitochondrial bioenergetics was measured at 72 h to capture the potential and accumulative effects of prolonged DEP exposure on mitochondrial function. Results: DEP profoundly altered mitochondrial morphology and network integrity, reduced both basal and ATP-linked respiration as well as the glycolytic capacity of mitochondria. DEP exposure increased gene expression of oxidative stress and inflammation markers such as interleukin-8 and interleukin-6. DEP significantly affected mRNA levels of exchange protein directly activated by cAMP-1 and -2 (Epac1, Epac2), appeared to increase Epac1 protein, but left phospho-PKA levels unhanged. DEP exposure increased A-kinase anchoring protein 1, β2-adrenoceptor and prostanoid E receptor subtype 4 mRNA levels. Interestingly, DEP decreased mRNA levels of adenylyl cyclase 9 and reduced cAMP levels stimulated by forskolin (AC activator), fenoterol (β2-AR agonist) or PGE2 (EPR agonist). Discussion: Our findings suggest that DEP induces mitochondrial dysfunction, a process accompanied by oxidative stress and inflammation, and broadly dampens cAMP signaling. These epithelial responses may contribute to lung dysfunction induced by air pollution exposure.
Collapse
Affiliation(s)
- Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ana L. Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hoeke A. Baarsma
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Asmaa Oun
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | | | - Emily Oosterhout
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rennolds S. Ostrom
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Samuel Santos Valenca
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
6
|
Gerde P, Sjöberg CO, Bäckroos H, Englund J, Wangheim M, Litorp H. Regional lung targeting with a fluticasone/salmeterol aerosol using a bolus breath hold method of the PreciseInhale® system: A first evaluation in humans. Eur J Pharm Sci 2024; 196:106742. [PMID: 38460609 DOI: 10.1016/j.ejps.2024.106742] [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: 09/06/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND In development of inhaled drugs- and formulations the measured concentration in the systemic circulation is often used as a surrogate for local dosimetry in the lungs. To further elucidate regional differences in the fate of drugs in the lungs, different aerodynamic sizes of aerosols have been used to target major airway regions. An alternative approach to achieve regional targeting of aerosols, is to use a defined aerosol bolus together with a bolus breath hold strategy. A small volume of test aerosol is intercalated and stopped at different penetration depths, to achieve increased drug deposition at chosen lung locations. Drug permeation from the lung regions is then investigated by repeatedly sampling venous blood from the systemic circulation. The PreciseInhale® (PI) exposure platform was developed to allow generation of aerosols from different sources, including clinical inhalers, into a holding chamber, for subsequent use with alternative exposure modules in vitro and in vivo. In the current first-in-human study was investigated the feasibility of a new clinical exposure module added to the PI system. By extracting aerosol puffs from a medical inhaler for subsequent delivery to volunteers, it was possible to administer whole lung exposures, as well as regional targeting exposures. METHODS Aerosols containing 250 µg/25 µg fluticasone propionate (FP)/salmeterol xinafoate (SMX) were automatically actuated and extracted from the pressurized Metered Dose Inhaler (pMDI) Evohaler Seretide forte into the PI system's holding chamber, then administered to the healthy volunteers using controlled flowrate and volume exposure cycles. Two main comparisons were made by measuring the systemic PK response: I. One label dose directly from the inhaler to the subject was compared to the same dose extracted from the pMDI into the PI system and then administered to the subject. II A small aerosol bolus at a penetration level in the central airways was compared to a small aerosol bolus at a penetration level in the peripheral lung. RESULTS AND CONCLUSIONS When one inhaler dose was administered via the PI system, the absorbed dose, expressed as AUC24, was approximately twice as high and the CV was less than half, compared to direct inhalation from the same pMDI. Bolus breath hold targeting of drugs from the same aerosol mixture to the peripheral lung and the central airways showed a difference in their appearance in the systemic circulation. Normalized to the same deposited dose, SMX had a 57 % higher Cmax in the peripheral lung compared to the central airways. However, from 6 to 24 h after dosing the systemic concentrations of SMX from both regions were quite similar. FP had parallel concentrations curves with a 23 % higher AUC24 in the peripheral lung with no noticeable elevation around Cmax. The permeability of these two substances from similar sized aerosols was indeed higher in the thinner air/blood barriers of the peripheral lung compared to the central airways, but differences as measured on the venous side of the circulation were not dramatic. In conclusion, the PI system provided better control of actuation, aspiration, and dispensation of aerosols from the clinical inhaler and thereby delivered higher quality read outs of pharmacokinetic parameters such as tmax, Cmax, and AUC. Improved performance, using PI system, can likely also be employed for studying regional selectivity of other responses in the lungs, for use in drug development.
Collapse
Affiliation(s)
- Per Gerde
- Inhalation Sciences AB, Novum, Hälsovägen 7, Huddinge SE-141 57, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| | - Carl-Olof Sjöberg
- Inhalation Sciences AB, Novum, Hälsovägen 7, Huddinge SE-141 57, Sweden; Flexura AB, Vitmåravägen 50, Upplands Väsby SE-194 60, Sweden
| | - Helen Bäckroos
- Inhalation Sciences AB, Novum, Hälsovägen 7, Huddinge SE-141 57, Sweden
| | - Joakim Englund
- Clinical Trial Consultants AB, Dag Hammarskjölds väg 10B, Uppsala SE-752 37, Sweden
| | - Marit Wangheim
- Clinical Trial Consultants AB, Dag Hammarskjölds väg 10B, Uppsala SE-752 37, Sweden
| | - Helena Litorp
- Clinical Trial Consultants AB, Dag Hammarskjölds väg 10B, Uppsala SE-752 37, Sweden; Department of Global Public Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Women's and Children's Health, Uppsala University, Stockholm, Sweden
| |
Collapse
|
7
|
Jaber N, Billet S. How to use an in vitro approach to characterize the toxicity of airborne compounds. Toxicol In Vitro 2024; 94:105718. [PMID: 37871865 DOI: 10.1016/j.tiv.2023.105718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/25/2023]
Abstract
As part of the development of new approach methodologies (NAMs), numerous in vitro methods are being developed to characterize the potential toxicity of inhalable xenobiotics (gases, volatile organic compounds, polycyclic aromatic hydrocarbons, particulate matter, nanoparticles). However, the materials and methods employed are extremely diverse, and no single method is currently in use. Method standardization and validation would raise trust in the results and enable them to be compared. This four-part review lists and compares biological models and exposure methodologies before describing measurable biomarkers of exposure or effect. The first section emphasizes the importance of developing alternative methods to reduce, if not replace, animal testing (3R principle). The biological models presented are mostly to cultures of epithelial cells from the respiratory system, as the lungs are the first organ to come into contact with air pollutants. Monocultures or cocultures of primary cells or cell lines, as well as 3D organotypic cultures such as organoids, spheroids and reconstituted tissues, but also the organ(s) model on a chip are examples. The exposure methods for these biological models applicable to airborne compounds are submerged, intermittent, continuous either static or dynamic. Finally, within the restrictions of these models (i.e. relative tiny quantities, adhering cells), the mechanisms of toxicity and the phenotypic markers most commonly examined in models exposed at the air-liquid interface (ALI) are outlined.
Collapse
Affiliation(s)
- Nour Jaber
- UR4492, Unité de Chimie Environnementale et Interactions sur le Vivant, Université du Littoral Côte d'Opale, Dunkerque, France
| | - Sylvain Billet
- UR4492, Unité de Chimie Environnementale et Interactions sur le Vivant, Université du Littoral Côte d'Opale, Dunkerque, France.
| |
Collapse
|
8
|
Petpiroon N, Netkueakul W, Sukrak K, Wang C, Liang Y, Wang M, Liu Y, Li Q, Kamran R, Naruse K, Aueviriyavit S, Takahashi K. Development of lung tissue models and their applications. Life Sci 2023; 334:122208. [PMID: 37884207 DOI: 10.1016/j.lfs.2023.122208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
The lungs are important organs that play a critical role in the development of specific diseases, as well as responding to the effects of drugs, chemicals, and environmental pollutants. Due to the ethical concerns around animal testing, alternative methods have been sought which are more time-effective, do not pose ethical issues for animals, do not involve species differences, and provide easy investigation of the pathobiology of lung diseases. Several national and international organizations are working to accelerate the development and implementation of structurally and functionally complex tissue models as alternatives to animal testing, particularly for the lung. Unfortunately, to date, there is no lung tissue model that has been accepted by regulatory agencies for use in inhalation toxicology. This review discusses the challenges involved in developing a relevant lung tissue model derived from human cells such as cell lines, primary cells, and pluripotent stem cells. It also introduces examples of two-dimensional (2D) air-liquid interface and monocultured and co-cultured three-dimensional (3D) culture techniques, particularly organoid culture and 3D bioprinting. Furthermore, it reviews development of the lung-on-a-chip model to mimic the microenvironment and physiological performance. The applications of lung tissue models in various studies, especially disease modeling, viral respiratory infection, and environmental toxicology will be also introduced. The development of a relevant lung tissue model is extremely important for standardizing and validation the in vitro models for inhalation toxicity and other studies in the future.
Collapse
Affiliation(s)
- Nalinrat Petpiroon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Woranan Netkueakul
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Kanokwan Sukrak
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand; Department of Environmental Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; Thailand Network Center on Air Quality Management: TAQM, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chen Wang
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Yin Liang
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Mengxue Wang
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Yun Liu
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Qiang Li
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Rumaisa Kamran
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Keiji Naruse
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan
| | - Sasitorn Aueviriyavit
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), 111 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand.
| | - Ken Takahashi
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ward, Okayama 700-8558, Japan.
| |
Collapse
|
9
|
Graf J, Trautmann-Rodriguez M, Sabnis S, Kloxin AM, Fromen CA. On the path to predicting immune responses in the lung: Modeling the pulmonary innate immune system at the air-liquid interface (ALI). Eur J Pharm Sci 2023; 191:106596. [PMID: 37770004 PMCID: PMC10658361 DOI: 10.1016/j.ejps.2023.106596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/01/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Chronic respiratory diseases and infections are among the largest contributors to death globally, many of which still have no cure, including chronic obstructive pulmonary disorder, idiopathic pulmonary fibrosis, and respiratory syncytial virus among others. Pulmonary therapeutics afford untapped potential for treating lung infection and disease through direct delivery to the site of action. However, the ability to innovate new therapeutic paradigms for respiratory diseases will rely on modeling the human lung microenvironment and including key cellular interactions that drive disease. One key feature of the lung microenvironment is the air-liquid interface (ALI). ALI interface modeling techniques, using cell-culture inserts, organoids, microfluidics, and precision lung slices (PCLS), are rapidly developing; however, one major component of these models is lacking-innate immune cell populations. Macrophages, neutrophils, and dendritic cells, among others, represent key lung cell populations, acting as the first responders during lung infection or injury. Innate immune cells respond to and modulate stromal cells and bridge the gap between the innate and adaptive immune system, controlling the bodies response to foreign pathogens and debris. In this article, we review the current state of ALI culture systems with a focus on innate immune cells and suggest ways to build on current models to add complexity and relevant immune cell populations.
Collapse
Affiliation(s)
- Jodi Graf
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - Simone Sabnis
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
10
|
Fang ZF, Fu Y, Peng Y, Song SR, Wang ZN, Yang Y, Nie YC, Han HL, Teng YB, Xiao WM, Chen JP, Zhou BJ, Ou GL, Xie JX, Liu XY, Zhang JJ, Zhong NS. Citrus peel extract protects against diesel exhaust particle-induced chronic obstructive pulmonary disease-like lung lesions and oxidative stress. Food Funct 2023; 14:9841-9856. [PMID: 37850547 DOI: 10.1039/d3fo02010j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide and characterized by emphysema, small airway remodeling and mucus hypersecretion. Citrus peels have been widely used as food spices and in traditional Chinese medicine for chronic lung disease. Given that citrus peels are known for containing antioxidants and anti-inflammatory compounds, we hypothesize that citrus peel intake can suppress oxidative stress and inflammatory response to air pollution exposure, thereby alleviating COPD-like pathologies. This study aimed to investigate the efficacy of citrus peel extract, namely Guang Chenpi (GC), in preventing the development of COPD induced by diesel exhaust particles (DEPs) and its potential mechanism. DEP-induced COPD-like lung pathologies, inflammatory responses and oxidative stress with or without GC treatment were examined in vivo and in vitro. Our in vivo study showed that GC was effective in decreasing inflammatory cell counts and inflammatory mediator (IL-17A and TNF-α) concentrations in bronchoalveolar lavage fluid (BALF). Pretreatment with GC extract also significantly decreased oxidative stress in the serum and lung tissue of DEP-induced COPD rats. Furthermore, GC pretreatment effectively reduced goblet cell hyperplasia (PAS positive cells) and fibrosis of the small airways, decreased macrophage infiltration as well as carbon loading in the peripheral lungs, and facilitated the resolution of emphysema and small airway remodeling in DEP-induced COPD rats. An in vitro free radical scavenging assay revealed robust antioxidant potential of GC in scavenging DPPH free radicals. Moreover, GC demonstrated potent capacities in reducing ROS production and enhancing SOD activity in BEAS-2B cells stimulated by DEPs. GC treatment significantly attenuated the increased level of IL-8 and MUC5AC from DEP-treated BEAS-2B cells. Mechanistically, GC treatment upregulated the protein level of Nrf-2 and could function via MAPK/NF-κB signaling pathways by suppressing the phosphorylation of p38, JNK and p65. Citrus peel extract is effective in decreasing oxidative stress and inflammatory responses of the peripheral lungs to DEP exposure. These protective effects further contributed to the resolution of COPD-like pathologies.
Collapse
Affiliation(s)
- Zhang-Fu Fang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen 518020, China
- Guangzhou Laboratory, Guangzhou 510320, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yu Fu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Sheng-Ren Song
- Department of Respiratory Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550001, China
| | - Zhao-Ni Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yang Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Yi-Chu Nie
- Translational Medicine Research Institute, First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Hai-Long Han
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Yan-Bo Teng
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Wei-Min Xiao
- Shenzhen Academy of Metrology & Quality Inspection, Shenzhen 518055, China
| | - Jia-Ping Chen
- Shenzhen Academy of Metrology & Quality Inspection, Shenzhen 518055, China
| | | | - Guo-Liang Ou
- Jiangmen Palace International Food, Inc., Jiangmen 529000, China
| | - Jia-Xing Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| | - Xiao-Yu Liu
- State Key Laboratory of Respiratory Disease Allergy Division at Shenzhen University, Institute of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen 518061, China.
| | - Junfeng Jim Zhang
- Nicholas School of the Environment and Global Health Institute, Duke University, Durham, North Carolina 27708, USA
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu 215316, China.
| | - Nan-Shan Zhong
- Guangzhou Laboratory, Guangzhou 510320, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
11
|
Gutor SS, Miller RF, Blackwell TS, Polosukhin VV. Environmental and occupational bronchiolitis obliterans: new reality. EBioMedicine 2023; 95:104760. [PMID: 37598462 PMCID: PMC10458287 DOI: 10.1016/j.ebiom.2023.104760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/10/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Patients diagnosed with environmental/occupational bronchiolitis obliterans (BO) over the last 2 decades often present with an indolent evolution of respiratory symptoms without a history of high-level, acute exposure to airborne toxins. Exertional dyspnea is the most common symptom and standard clinical and radiographic evaluation can be non-diagnostic. Lung biopsies often reveal pathological abnormalities affecting all distal lung compartments. These modern cases of BO typically exhibit the constrictive bronchiolitis phenotype of small airway remodeling, along with lymphocytic inflammation. In addition, hypertensive-type remodeling of intrapulmonary vasculature, diffuse fibroelastosis of alveolar tissue, and fibrous thickening of visceral pleura are frequently present. The diagnosis of environmental/occupational BO should be considered in patients who present with subacute onset of exertional dyspnea and a history compatible with prolonged or recurrent exposure to environmental toxins. Important areas for future studies include development of less invasive diagnostic approaches and testing of novel agents for disease prevention and treatment.
Collapse
Affiliation(s)
- Sergey S Gutor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert F Miller
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Medical Center, Nashville, TN, USA
| | - Vasiliy V Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
12
|
Rahman M, Upadhyay S, Ganguly K, Introna M, Ji J, Boman C, Muala A, Blomberg A, Sandström T, Palmberg L. Comparable Response Following Exposure to Biodiesel and Diesel Exhaust Particles in Advanced Multicellular Human Lung Models. TOXICS 2023; 11:532. [PMID: 37368632 DOI: 10.3390/toxics11060532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Biodiesel is considered to be a sustainable alternative for fossil fuels such as petroleum-based diesel. However, we still lack knowledge about the impact of biodiesel emissions on humans, as airways and lungs are the primary target organs of inhaled toxicants. This study investigated the effect of exhaust particles from well-characterized rapeseed methyl ester (RME) biodiesel exhaust particles (BDEP) and petro-diesel exhaust particles (DEP) on primary bronchial epithelial cells (PBEC) and macrophages (MQ). The advanced multicellular physiologically relevant bronchial mucosa models were developed using human primary bronchial epithelial cells (PBEC) cultured at air-liquid interface (ALI) in the presence or absence of THP-1 cell-derived macrophages (MQ). The experimental set-up used for BDEP and DEP exposures (18 µg/cm2 and 36 µg/cm2) as well as the corresponding control exposures were PBEC-ALI, MQ-ALI, and PBEC co-cultured with MQ (PBEC-ALI/MQ). Following exposure to both BDEP and DEP, reactive oxygen species as well as the stress protein heat shock protein 60 were upregulated in PBEC-ALI and MQ-ALI. Expression of both pro-inflammatory (M1: CD86) and repair (M2: CD206) macrophage polarization markers was increased in MQ-ALI after both BDEP and DEP exposures. Phagocytosis activity of MQ and the phagocytosis receptors CD35 and CD64 were downregulated, whereas CD36 was upregulated in MQ-ALI. Increased transcript and secreted protein levels of CXCL8, as well as IL-6 and TNF-α, were detected following both BDEP and DEP exposure at both doses in PBEC-ALI. Furthermore, the cyclooxygenase-2 (COX-2) pathway, COX-2-mediated histone phosphorylation and DNA damage were all increased in PBEC-ALI following exposure to both doses of BDEP and DEP. Valdecoxib, a COX-2 inhibitor, reduced the level of prostaglandin E2, histone phosphorylation, and DNA damage in PBEC-ALI following exposure to both concentrations of BDEP and DEP. Using physiologically relevant multicellular human lung mucosa models with human primary bronchial epithelial cells and macrophages, we found BDEP and DEP to induce comparable levels of oxidative stress, inflammatory response, and impairment of phagocytosis. The use of a renewable carbon-neutral biodiesel fuel does not appear to be more favorable than conventional petroleum-based alternative, as regards of its potential for adverse health effects.
Collapse
Affiliation(s)
- Mizanur Rahman
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Micol Introna
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Jie Ji
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Christoffer Boman
- Thermochemical Energy Conversion Laboratory, Department of Applied Physics and Electronics, Umeå University, 901 87 Umeå, Sweden
| | - Ala Muala
- Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Anders Blomberg
- Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Umeå University, 901 87 Umeå, Sweden
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
13
|
Al-Rekabi Z, Dondi C, Faruqui N, Siddiqui NS, Elowsson L, Rissler J, Kåredal M, Mudway I, Larsson-Callerfelt AK, Shaw M. Uncovering the cytotoxic effects of air pollution with multi-modal imaging of in vitro respiratory models. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221426. [PMID: 37063998 PMCID: PMC10090883 DOI: 10.1098/rsos.221426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/17/2023] [Indexed: 06/19/2023]
Abstract
Annually, an estimated seven million deaths are linked to exposure to airborne pollutants. Despite extensive epidemiological evidence supporting clear associations between poor air quality and a range of short- and long-term health effects, there are considerable gaps in our understanding of the specific mechanisms by which pollutant exposure induces adverse biological responses at the cellular and tissue levels. The development of more complex, predictive, in vitro respiratory models, including two- and three-dimensional cell cultures, spheroids, organoids and tissue cultures, along with more realistic aerosol exposure systems, offers new opportunities to investigate the cytotoxic effects of airborne particulates under controlled laboratory conditions. Parallel advances in high-resolution microscopy have resulted in a range of in vitro imaging tools capable of visualizing and analysing biological systems across unprecedented scales of length, time and complexity. This article considers state-of-the-art in vitro respiratory models and aerosol exposure systems and how they can be interrogated using high-resolution microscopy techniques to investigate cell-pollutant interactions, from the uptake and trafficking of particles to structural and functional modification of subcellular organelles and cells. These data can provide a mechanistic basis from which to advance our understanding of the health effects of airborne particulate pollution and develop improved mitigation measures.
Collapse
Affiliation(s)
- Zeinab Al-Rekabi
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Camilla Dondi
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Nilofar Faruqui
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
| | - Nazia S. Siddiqui
- Faculty of Medical Sciences, University College London, London, UK
- Kingston Hospital NHS Foundation Trust, Kingston upon Thames, UK
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jenny Rissler
- Bioeconomy and Health, RISE Research Institutes of Sweden, Lund, Sweden
- Ergonomics and Aerosol Technology, Lund University, Lund, Sweden
| | - Monica Kåredal
- Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Ian Mudway
- MRC Centre for Environment and Health, Imperial College London, London, UK
- National Institute of Health Protection Research Unit in Environmental Exposures and Health, London, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - Michael Shaw
- Department of Chemical and Biological Sciences, National Physical Laboratory, Teddington, UK
- Department of Computer Science, University College London, London, UK
| |
Collapse
|
14
|
Sengupta A, Dorn A, Jamshidi M, Schwob M, Hassan W, De Maddalena LL, Hugi A, Stucki AO, Dorn P, Marti TM, Wisser O, Stucki JD, Krebs T, Hobi N, Guenat OT. A multiplex inhalation platform to model in situ like aerosol delivery in a breathing lung-on-chip. Front Pharmacol 2023; 14:1114739. [PMID: 36959848 PMCID: PMC10029733 DOI: 10.3389/fphar.2023.1114739] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Prolonged exposure to environmental respirable toxicants can lead to the development and worsening of severe respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD) and fibrosis. The limited number of FDA-approved inhaled drugs for these serious lung conditions has led to a shift from in vivo towards the use of alternative in vitro human-relevant models to better predict the toxicity of inhaled particles in preclinical research. While there are several inhalation exposure models for the upper airways, the fragile and dynamic nature of the alveolar microenvironment has limited the development of reproducible exposure models for the distal lung. Here, we present a mechanistic approach using a new generation of exposure systems, the Cloud α AX12. This novel in vitro inhalation tool consists of a cloud-based exposure chamber (VITROCELL) that integrates the breathing AXLung-on-chip system (AlveoliX). The ultrathin and porous membrane of the AX12 plate was used to create a complex multicellular model that enables key physiological culture conditions: the air-liquid interface (ALI) and the three-dimensional cyclic stretch (CS). Human-relevant cellular models were established for a) the distal alveolar-capillary interface using primary cell-derived immortalized alveolar epithelial cells (AXiAECs), macrophages (THP-1) and endothelial (HLMVEC) cells, and b) the upper-airways using Calu3 cells. Primary human alveolar epithelial cells (AXhAEpCs) were used to validate the toxicity results obtained from the immortalized cell lines. To mimic in vivo relevant aerosol exposures with the Cloud α AX12, three different models were established using: a) titanium dioxide (TiO2) and zinc oxide nanoparticles b) polyhexamethylene guanidine a toxic chemical and c) an anti-inflammatory inhaled corticosteroid, fluticasone propionate (FL). Our results suggest an important synergistic effect on the air-blood barrier sensitivity, cytotoxicity and inflammation, when air-liquid interface and cyclic stretch culture conditions are combined. To the best of our knowledge, this is the first time that an in vitro inhalation exposure system for the distal lung has been described with a breathing lung-on-chip technology. The Cloud α AX12 model thus represents a state-of-the-art pre-clinical tool to study inhalation toxicity risks, drug safety and efficacy.
Collapse
Affiliation(s)
- Arunima Sengupta
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Aurélien Dorn
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Mohammad Jamshidi
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Magali Schwob
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Widad Hassan
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | | | - Andreas Hugi
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Andreas O. Stucki
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- *Correspondence: Andreas O. Stucki,
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Thomas M. Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | | | - Nina Hobi
- AlveoliX AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Olivier T. Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
15
|
Bannuscher A, Schmid O, Drasler B, Rohrbasser A, Braakhuis HM, Meldrum K, Zwart EP, Gremmer ER, Birk B, Rissel M, Landsiedel R, Moschini E, Evans SJ, Kumar P, Orak S, Doryab A, Erdem JS, Serchi T, Vandebriel RJ, Cassee FR, Doak SH, Petri-Fink A, Zienolddiny S, Clift MJD, Rothen-Rutishauser B. An inter-laboratory effort to harmonize the cell-delivered in vitro dose of aerosolized materials. NANOIMPACT 2022; 28:100439. [PMID: 36402283 DOI: 10.1016/j.impact.2022.100439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
Air-liquid interface (ALI) lung cell models cultured on permeable transwell inserts are increasingly used for respiratory hazard assessment requiring controlled aerosolization and deposition of any material on ALI cells. The approach presented herein aimed to assess the transwell insert-delivered dose of aerosolized materials using the VITROCELL® Cloud12 system, a commercially available aerosol-cell exposure system. An inter-laboratory comparison study was conducted with seven European partners having different levels of experience with the VITROCELL® Cloud12. A standard operating procedure (SOP) was developed and applied by all partners for aerosolized delivery of materials, i.e., a water-soluble molecular substance (fluorescence-spiked salt) and two poorly soluble particles, crystalline silica quartz (DQ12) and titanium dioxide nanoparticles (TiO2 NM-105). The material dose delivered to transwell inserts was quantified with spectrofluorometry (fluorescein) and with the quartz crystal microbalance (QCM) integrated in the VITROCELL® Cloud12 system. The shape and agglomeration state of the deposited particles were confirmed with transmission electron microscopy (TEM). Inter-laboratory comparison of the device-specific performance was conducted in two steps, first for molecular substances (fluorescein-spiked salt), and then for particles. Device- and/or handling-specific differences in aerosol deposition of VITROCELL® Cloud12 systems were characterized in terms of the so-called deposition factor (DF), which allows for prediction of the transwell insert-deposited particle dose from the particle concentration in the aerosolized suspension. Albeit DF varied between the different labs from 0.39 to 0.87 (mean (coefficient of variation (CV)): 0.64 (28%)), the QCM of each VITROCELL® Cloud 12 system accurately measured the respective transwell insert-deposited dose. Aerosolized delivery of DQ12 and TiO2 NM-105 particles showed good linearity (R2 > 0.95) between particle concentration of the aerosolized suspension and QCM-determined insert-delivered particle dose. The VITROCELL® Cloud 12 performance for DQ12 particles was identical to that for fluorescein-spiked salt, i.e., the ratio of measured and salt-predicted dose was 1.0 (29%). On the other hand, a ca. 2-fold reduced dose was observed for TiO2 NM-105 (0.54 (41%)), which was likely due to partial retention of TiO2 NM-105 agglomerates in the vibrating mesh nebulizer of the VITROCELL® Cloud12. This inter-laboratory comparison demonstrates that the QCM integrated in the VITROCELL® Cloud 12 is a reliable tool for dosimetry, which accounts for potential variations of the transwell insert-delivered dose due to device-, handling- and/or material-specific effects. With the detailed protocol presented herein, all seven partner laboratories were able to demonstrate dose-controlled aerosolization of material suspensions using the VITROCELL® Cloud12 exposure system at dose levels relevant for observing in vitro hazard responses. This is an important step towards regulatory approved implementation of ALI lung cell cultures for in vitro hazard assessment of aerosolized materials.
Collapse
Affiliation(s)
- Anne Bannuscher
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC-M), Helmholtz Zentrum München - Member of the German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany; Institute of Lung Health and Immunity, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Alain Rohrbasser
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Hedwig M Braakhuis
- National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Kirsty Meldrum
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Medical School, Institute of Life Sciences, Centre for NanoHealth, Swansea University, Singleton Campus, Wales SA2 8PP, UK
| | - Edwin P Zwart
- National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Eric R Gremmer
- National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Barbara Birk
- BASF SE, Experimental Toxicology and Ecology, 67056 Ludwigshafen am Rhein, Germany
| | - Manuel Rissel
- BASF SE, Experimental Toxicology and Ecology, 67056 Ludwigshafen am Rhein, Germany
| | - Robert Landsiedel
- BASF SE, Experimental Toxicology and Ecology, 67056 Ludwigshafen am Rhein, Germany; Free University of Berlin, Pharmacy, Pharmacology and Toxicology, 14195 Berlin, Germany
| | - Elisa Moschini
- Department of Environmental Research and Innovation, Luxembourg Institute of Science and Technology (LIST), 41 rue du Brill, L4422 Belvaux, Grand-Duchy of Luxembourg, Luxembourg
| | - Stephen J Evans
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Medical School, Institute of Life Sciences, Centre for NanoHealth, Swansea University, Singleton Campus, Wales SA2 8PP, UK
| | - Pramod Kumar
- Comprehensive Pneumology Center (CPC-M), Helmholtz Zentrum München - Member of the German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany; Institute of Lung Health and Immunity, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Sezer Orak
- Comprehensive Pneumology Center (CPC-M), Helmholtz Zentrum München - Member of the German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany; Institute of Lung Health and Immunity, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Ali Doryab
- Comprehensive Pneumology Center (CPC-M), Helmholtz Zentrum München - Member of the German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany; Institute of Lung Health and Immunity, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Tommaso Serchi
- Department of Environmental Research and Innovation, Luxembourg Institute of Science and Technology (LIST), 41 rue du Brill, L4422 Belvaux, Grand-Duchy of Luxembourg, Luxembourg
| | - Rob J Vandebriel
- National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Flemming R Cassee
- National Institute for Public Health and the Environment (RIVM), PO Box 1, 3720, BA, Bilthoven, the Netherlands; Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Shareen H Doak
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Medical School, Institute of Life Sciences, Centre for NanoHealth, Swansea University, Singleton Campus, Wales SA2 8PP, UK
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | | | - Martin J D Clift
- In Vitro Toxicology Group, Faculty of Medicine, Health and Life Sciences, Medical School, Institute of Life Sciences, Centre for NanoHealth, Swansea University, Singleton Campus, Wales SA2 8PP, UK
| | | |
Collapse
|
16
|
Rahman M, Irmler M, Introna M, Beckers J, Palmberg L, Johanson G, Upadhyay S, Ganguly K. Insight into the pulmonary molecular toxicity of heated tobacco products using human bronchial and alveolar mucosa models at air-liquid interface. Sci Rep 2022; 12:16396. [PMID: 36180488 PMCID: PMC9525689 DOI: 10.1038/s41598-022-20657-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/16/2022] [Indexed: 11/09/2022] Open
Abstract
Heated tobacco products (HTP) are novel nicotine delivery products with limited toxicological data. HTP uses heating instead of combustion to generate aerosol (HTP-smoke). Physiologically relevant human bronchial and alveolar lung mucosa models developed at air-liquid interface were exposed to HTP-smoke to assess broad toxicological response (n = 6-7; ISO puffing regimen; compared to sham; non-parametric statistical analysis; significance: p < 0.05). Elevated levels of total cellular reactive oxygen species, stress responsive nuclear factor kappa-B, and DNA damage markers [8-hydroxy-2'-deoxyguanosine, phosphorylated histone H2AX, cleaved poly-(ADP-Ribose) polymerase] were detected in HTP-smoke exposed bronchial and/or alveolar models. RNA sequencing detected differential regulation of 724 genes in the bronchial- and 121 genes in the alveolar model following HTP-smoke exposure (cut off: p ≤ 0.01; fold change: ≥ 2). Common enriched pathways included estrogen biosynthesis, ferroptosis, superoxide radical degradation, xenobiotics, and α-tocopherol degradation. Secreted levels of interleukin (IL)1ꞵ and IL8 increased in the bronchial model whereas in the alveolar model, interferon-γ and IL4 increased and IL13 decreased following HTP-smoke exposure. Increased lipid peroxidation was detected in HTP-smoke exposed bronchial and alveolar models which was inhibited by ferrostatin-1. The findings form a basis to perform independent risk assessment studies on different flavours of HTP using different puffing topography and corresponding chemical characterization.
Collapse
Affiliation(s)
- Mizanur Rahman
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764, Neuherberg, Germany
| | - Micol Introna
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD E.V.), 85764, Neuherberg, Germany
- Chair of Experimental Genetics, Technical University of Munich, 85354, Freising, Germany
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Gunnar Johanson
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
17
|
Makutani Y, Kawakami H, Tsujikawa T, Yoshimura K, Chiba Y, Ito A, Kawamura J, Haratani K, Nakagawa K. Contribution of MMP14-expressing cancer-associated fibroblasts in the tumor immune microenvironment to progression of colorectal cancer. Front Oncol 2022; 12:956270. [PMID: 36052235 PMCID: PMC9424903 DOI: 10.3389/fonc.2022.956270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Matrix metalloproteinase 14 (MMP14) expression is implicated in progression of colorectal cancer, but its role in the tumor microenvironment (TME) has been unclear. The relevance of MMP14 to colorectal cancer progression was explored by analysis of transcriptomic data for colorectal adenocarcinoma patients (n = 592) in The Cancer Genome Atlas. The role of MMP14 in the TME was investigated in a retrospective analysis of tumor samples from 86 individuals with stage III colorectal cancer by single cell–based spatial profiling of MMP14 expression as performed by 12-color multiplex immunohistochemistry (mIHC). Analysis of gene expression data revealed that high MMP14 expression was associated with tumor progression and implicated both cancer-associated fibroblasts (CAFs) and tumor-associated macrophages in such progression. Spatial profiling by mIHC revealed that a higher percentage of MMP14+ cells among intratumoral CAFs (MMP14+ CAF/CAF ratio) was associated with poorer relapse-free survival. Multivariable analysis including key clinical factors identified the MMP14+ CAF/CAF ratio as an independent poor prognostic factor. Moreover, the patient subset with both a high MMP14+ CAF/CAF ratio and a low tumor-infiltrating lymphocyte density showed the worst prognosis. Our results suggest that MMP14+ CAFs play an important role in progression of stage III colorectal cancer and may therefore be a promising therapeutic target.
Collapse
Affiliation(s)
- Yusuke Makutani
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- *Correspondence: Hisato Kawakami, ; Koji Haratani,
| | - Takahiro Tsujikawa
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanako Yoshimura
- Department of Otolaryngology–Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka-Sayama, Japan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Junichiro Kawamura
- Department of Surgery, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
- *Correspondence: Hisato Kawakami, ; Koji Haratani,
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
18
|
Upadhyay S, Chakraborty A, Thimraj TA, Baldi M, Steneholm A, Ganguly K, Gerde P, Ernstgård L, Palmberg L. Establishment of Repeated In Vitro Exposure System for Evaluating Pulmonary Toxicity of Representative Criteria Air Pollutants Using Advanced Bronchial Mucosa Models. TOXICS 2022; 10:toxics10060277. [PMID: 35736886 PMCID: PMC9228979 DOI: 10.3390/toxics10060277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
There is mounting evidence that shows the association between chronic exposure to air pollutants (particulate matter and gaseous) and onset of various respiratory impairments. However, the corresponding toxicological mechanisms of mixed exposure are poorly understood. Therefore, in this study, we aimed to establish a repeated exposure setting for evaluating the pulmonary toxicological effects of diesel exhaust particles (DEP), nitrogen dioxide (NO2), and sulfur dioxide (SO2) as representative criterial air pollutants. Single, combined (DEP with NO2 and SO2), and repeated exposures were performed using physiologically relevant human bronchial mucosa models developed at the air−liquid interface (bro-ALI). The bro-ALI models were generated using human primary bronchial epithelial cells (3−4 donors; 2 replicates per donor). The exposure regime included the following: 1. DEP (12.5 µg/cm2; 3 min/day, 3 days); 2. low gaseous (NO2: 0.1 ppm + SO2: 0.2 ppm); (30 min/day, 3 days); 3. high gaseous (NO2: 0.2 ppm + SO2: 0.4 ppm) (30 min/day, 3 days); and 4. single combined (DEP + low gaseous for 1 day). The markers for pro-inflammatory (IL8, IL6, NFKB, TNF), oxidative stress (HMOX1, GSTA1, SOD3,) and tissue injury/repair (MMP9, TIMP1) responses were assessed at transcriptional and/ or secreted protein levels following exposure. The corresponding sham-exposed samples under identical conditions served as the control. A non-parametric statistical analysis was performed and p < 0.05 was considered as significant. Repeated exposure to DEP and single combined (DEP + low gaseous) exposure showed significant alteration in the pro-inflammatory, oxidative stress and tissue injury responses compared to repeated exposures to gaseous air pollutants. The study demonstrates that it is feasible to predict the long-term effects of air pollutants using the above explained exposure system.
Collapse
Affiliation(s)
- Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
- Correspondence: (S.U.); (L.P.); Tel.:+46-85-2487930 (S.U.); +46-8-524-822-10 (L.P.)
| | - Ashesh Chakraborty
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Tania A. Thimraj
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Marialuisa Baldi
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | | | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Per Gerde
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Lena Ernstgård
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
- Correspondence: (S.U.); (L.P.); Tel.:+46-85-2487930 (S.U.); +46-8-524-822-10 (L.P.)
| |
Collapse
|
19
|
Zerboni A, Rossi T, Bengalli R, Catelani T, Rizzi C, Priola M, Casadei S, Mantecca P. Diesel exhaust particulate emissions and in vitro toxicity from Euro 3 and Euro 6 vehicles. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118767. [PMID: 34974087 DOI: 10.1016/j.envpol.2021.118767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Incomplete combustion processes in diesel engines produce particulate matter (PM) that significantly contributes to air pollution. Currently, there remains a knowledge gap in relation to the physical and chemical characteristics and also the biological reactivity of the PM emitted from old- and new-generation diesel vehicles. In this study, the emissions from a Euro 3 diesel vehicle were compared to those from a Euro 6 car during the regeneration of a diesel particulate filter (DPF). Different driving cycles were used to collect two types of diesel exhaust particles (DEPs). The particle size distribution was monitored using an engine exhaust particle sizer spectrometer and an electrical low-pressure impactor. Although the Euro 6 vehicle emitted particulates only during DPF regeneration that primarily occurs for a few minutes at high speeds, such emissions are characterized by a higher number of ultrafine particles (<0.1 μm) compared to those from the Euro 3 diesel vehicle. The emitted particles possess different characteristics. For example, Euro 6 DEPs exhibit a lower PAH content than do Euro 3 samples; however, they are enriched in metals that were poorly detected or undetected in Euro 3 emissions. The biological effects of the two DEPs were investigated in human bronchial BEAS-2B cells exposed to 50 μg/mL of PM (corresponding to 5.2 μg/cm2), and the results revealed that Euro 3 DEPs activated the typical inflammatory and pro-carcinogenic pathways induced by combustion-derived particles, while Euro 6 DEPs were less effective in regard to activating such biological responses. Although further investigations are required, it is evident that the different in vitro effects elicited by Euro 3 and Euro 6 DEPs can be correlated with the variable chemical compositions (metals and PAHs) of the emitted particles that play a pivotal role in the inflammatory and carcinogenic potential of airborne PM.
Collapse
Affiliation(s)
- Alessandra Zerboni
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy.
| | - Tommaso Rossi
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Rossella Bengalli
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| | - Tiziano Catelani
- Microscopy Facility, University of Milano-Bicocca, Piazza della Scienza 3, 20126, Milano, Italy
| | - Cristiana Rizzi
- Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| | - Marco Priola
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Simone Casadei
- Innovhub-SSI Fuels Department, Via Galileo Galilei, 1, 20097, San Donato Milanese, Milan, Italy
| | - Paride Mantecca
- Polaris Research Centre, Dept. of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza, 1, 20126, Milan, Italy
| |
Collapse
|
20
|
Sun Y, Kinsela AS, Cen X, Sun S, Collins RN, Cliff DI, Wu Y, Waite TD. Impact of reactive iron in coal mine dust on oxidant generation and epithelial lung cell viability. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 810:152277. [PMID: 34902414 DOI: 10.1016/j.scitotenv.2021.152277] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/25/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
Coal workers' pneumoconiosis (CWP) is a preventable occupational lung disease caused by the chronic inhalation of coal mine dust. The inhalation of coal mine dusts can result in the development of a range of lung diseases termed coal mine dust lung diseases, which is not limited to CWP and includes silicosis, bronchitis, emphysema and cancer. For decades, the presence of elemental Fe, C and Si has been proposed to be the causal factors underlying CWP. The recent resurgence of CWP globally with examination of cases in the United States suggesting a potential but inconclusive role of Fe(II)-sulfide minerals. To obtain a better understanding of Australian coals, the existence and potential adverse impacts of iron minerals were examined using 24 representative Australian coal samples. The results of this work revealed that reduced iron minerals were widely distributed within samples obtained from Australian coal mines with pyrite and siderite being particularly abundant. Compared with carbon and crystalline silica, the presence of these specific iron minerals were negatively correlated to the viability of both alveolar macrophages (NR8383) and human lung epithelial cells (A549) (R2 = 0.689) under scenarios reflecting biologically-relevant inflammatory response conditions. Further analysis using Welch's unpaired t-test indicated that the presence of reduced iron minerals statistically enhanced acellular oxidant production (90% CI [0.74 to 2.55]) and inflammatory response (90% CI [0.15 to 36.96]). Compared with Fe(II)-hydroxide, Fe(II)- and Fe(III)-(phyllo)silicate and Fe(II)-sulfate mineralogies, pyrite and siderite bearing dusts are likely to have greater adverse impacts on epithelial lung cells under inflammatory response conditions in view of both their iron content and reactivity.
Collapse
Affiliation(s)
- Yingying Sun
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Andrew S Kinsela
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiaotong Cen
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Siqi Sun
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Richard N Collins
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - David I Cliff
- Minerals Industry Safety and Health Centre, Sustainable Minerals Institute, University of Queensland, Brisbane, St Lucia, QLD 4072, Australia
| | - Yuxuan Wu
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia
| | - T David Waite
- School of Civil and Environmental Engineering, Water Research Centre, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
21
|
Aryl Hydrocarbon Receptor (AhR) Limits the Inflammatory Responses in Human Lung Adenocarcinoma A549 Cells via Interference with NF-κB Signaling. Cells 2022; 11:cells11040707. [PMID: 35203356 PMCID: PMC8870046 DOI: 10.3390/cells11040707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/01/2022] [Accepted: 02/14/2022] [Indexed: 02/08/2023] Open
Abstract
Apart from its role in the metabolism of carcinogens, the aryl hydrocarbon receptor (AhR) has been suggested to be involved in the control of inflammatory responses within the respiratory tract. However, the mechanisms responsible for this are only partially known. In this study, we used A549 cell line, as a human model of lung alveolar type II (ATII)-like cells, to study the functional role of the AhR in control of inflammatory responses. Using IL-1β as an inflammation inducer, we found that the induction of cyclooxygenase-2 and secretion of prostaglandins, as well as expression and release of pro-inflammatory cytokines, were significantly higher in the AhR-deficient A549 cells. This was linked with an increased nuclear factor-κB (NF-κB) activity, and significantly enhanced phosphorylation of its regulators, IKKα/β, and their target IκBα, in the AhR-deficient A549 cells. In line with this, when we mimicked the exposure to a complex mixture of airborne pollutants, using an organic extract of reference diesel exhaust particle mixture, an exacerbated inflammatory response was observed in the AhR-deficient cells, as compared with wild-type A549 cells. Together, the present results indicate that the AhR may act as a negative regulator of the inflammatory response in the A549 model, via a direct modulation of NF-κB signaling. Its role(s) in the control of inflammation within the lung alveoli exposed to airborne pollutants, especially those which simultaneously activate the AhR, thus deserve further attention.
Collapse
|
22
|
Shen F, Li D, Chen J. Mechanistic toxicity assessment of fine particulate matter emitted from fuel combustion via pathway-based approaches in human cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150214. [PMID: 34571223 DOI: 10.1016/j.scitotenv.2021.150214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/03/2021] [Accepted: 09/03/2021] [Indexed: 06/13/2023]
Abstract
Fuel exhaust particulate matter (FEPM) is an important source of air pollution worldwide. However, the comparative and mechanistic toxicity of FEPMs emitted from combustion of different fuels is still not fully understood. This study employed pathway-based approaches via human cells to evaluate mechanistic toxicity of FEPMs. The results showed that FEPMs caused concentration-dependent (0.1-200 μg/mL) cytotoxicity and oxidative stress. FEPMs at low concentration (10 μg/mL) induced cell cycle arrest in S and G2 phases, while high level of FEPMs (200 μg/mL) caused cell cycle arrest in G1 phase. Different FEPMs induced distinct expression profiles of toxicity-related genes, illustrating different toxic mechanisms. Furthermore, FEPMs inhibited the phosphorylation of protein kinase A (PKA), which related with reproductive toxicity. Spearman rank correlations among the chemicals carried by FEPMs and the toxic effects revealed that PAHs and metals promoted cell cycle arrest in the G1 phase and suppressed PKA activity. Furthermore, PAHs (Nap and Acy) and metals (Al and Pb) in FEPMs were highly and positively correlated with the expression of genes involved in apoptosis, ER stress, metal stress and inflammation. Our findings offered more mechanistic information of FEPMs at the level of subcellular toxicity and help to better understand their potential health effects.
Collapse
Affiliation(s)
- Fanglin Shen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China; Fudan Tyndall Center, Department of Environmental Science & Engineering, Fudan University, Shanghai 200438, China
| | - Dan Li
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China.
| | - Jianmin Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution and Prevention (LAP(3)), IRDR ICoE on Risk Interconnectivity and Governance on Weather/Climate Extremes Impact and Public Health, Department of Environmental Science and Engineering, Fudan University, Shanghai 200433, China; Fudan Tyndall Center, Department of Environmental Science & Engineering, Fudan University, Shanghai 200438, China
| |
Collapse
|
23
|
He RW, Houtzager MMG, Jongeneel WP, Westerink RHS, Cassee FR. In vitro hazard characterization of simulated aircraft cabin bleed-air contamination in lung models using an air-liquid interface (ALI) exposure system. ENVIRONMENT INTERNATIONAL 2021; 156:106718. [PMID: 34166876 DOI: 10.1016/j.envint.2021.106718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 06/13/2023]
Abstract
Contamination of aircraft cabin air can result from leakage of engine oils and hydraulic fluids into bleed air. This may cause adverse health effects in cabin crews and passengers. To realistically mimic inhalation exposure to aircraft cabin bleed-air contaminants, a mini bleed-air contaminants simulator (Mini-BACS) was constructed and connected to an air-liquid interface (ALI) aerosol exposure system (AES). This unique "Mini-BACS + AES" setup provides steady conditions to perform ALI exposure of the mono- and co-culture lung models to fumes from pyrolysis of aircraft engine oils and hydraulic fluids at respectively 200 °C and 350 °C. Meanwhile, physicochemical characteristics of test atmospheres were continuously monitored during the entire ALI exposure, including chemical composition, particle number concentration (PNC) and particles size distribution (PSD). Additional off-line chemical characterization was also performed for the generated fume. We started with submerged exposure to fumes generated from 4 types of engine oil (Fume A, B, C, and D) and 2 types of hydraulic fluid (Fume E and F). Following submerged exposures, Fume E and F as well as Fume A and B exerted the highest toxicity, which were therefore further tested under ALI exposure conditions. ALI exposures reveal that these selected engine oil (0-100 mg/m3) and hydraulic fluid (0-90 mg/m3) fumes at tested dose-ranges can impair epithelial barrier functions, induce cytotoxicity, produce pro-inflammatory responses, and reduce cell viability. Hydraulic fluid fumes are more toxic than engine oil fumes on the mass concentration basis. This may be related to higher abundance of organophosphates (OPs, ≈2800 µg/m3) and smaller particle size (≈50 nm) of hydraulic fluid fumes. Our results suggest that exposure to engine oil and hydraulic fluid fumes can induce considerable lung toxicity, clearly reflecting the potential health risks of contaminated aircraft cabin air.
Collapse
Affiliation(s)
- Rui-Wen He
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Toxicology Division, Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80177, 3508 TD Utrecht, the Netherlands
| | - Marc M G Houtzager
- The Netherlands Organisation for Applied Scientific Research, TNO, P.O. Box 80015, 3508 TA Utrecht, the Netherlands
| | - W P Jongeneel
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, the Netherlands
| | - Remco H S Westerink
- Institute for Risk Assessment Sciences (IRAS), Toxicology Division, Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80177, 3508 TD Utrecht, the Netherlands
| | - Flemming R Cassee
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720 BA Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Toxicology Division, Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80177, 3508 TD Utrecht, the Netherlands.
| |
Collapse
|
24
|
Müller L, Usemann J, Alves MP, Latzin P. Diesel exposure increases susceptibility of primary human nasal epithelial cells to rhinovirus infection. Physiol Rep 2021; 9:e14994. [PMID: 34542243 PMCID: PMC8451029 DOI: 10.14814/phy2.14994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/30/2022] Open
Abstract
Nasal epithelial cells (NECs) are among the first cells to be exposed to air pollutants and respiratory viruses. Although it is known that air pollution exposure and rhinovirus infections increase the risk for asthma development independently, it is unclear how these risk factors interact on a cellular level. Therefore, we aimed to investigate how exposure to diesel particulate matter (DPM) modifies the response of primary NECs to rhinovirus (RV) infection in vitro. Exposure of re-differentiated, primary NECs (49 healthy children [0-7 years], 12 adults) to DPM modified the mRNA expression of viral cell-surface receptors, pattern recognition receptors, and pro-inflammatory response (also protein levels). After exposure to DPM, we additionally infected the NECs with RV-1b and RV-16. Viral loads (assessed by titration assays) were significantly higher in DPM-exposed compared with non-exposed NECs. Exposure to DPM prior to RV infection resulted in a significant upregulation of pro-inflammatory cytokines (mRNA and protein level) and β-defensins mRNA, and significant downregulation of pattern recognition receptors mRNA and CXCL10 (mRNA and protein levels). There was no difference between all outcomes of NECs from children and adults. We can conclude that exposure to DPM prior to RV infection increases viral loads by downregulation of viral defense receptors and upregulation of pro-inflammatory cytokines. Our findings indicate a strong interaction between air pollution and the antiviral response to RV infection in NECs. We provide mechanistic evidence that exposure to air pollution increases susceptibility to RV infection.
Collapse
Affiliation(s)
- Loretta Müller
- Division of Paediatric Respiratory Medicine and AllergologyDepartment of Paediatrics, InselspitalBern University HospitalUniversity of BernBernSwitzerland
- Department for BioMedical Research (DBMR)University of BernBernSwitzerland
- University Children's Hospital Basel (UKBB)BaselSwitzerland
| | - Jakob Usemann
- Division of Paediatric Respiratory Medicine and AllergologyDepartment of Paediatrics, InselspitalBern University HospitalUniversity of BernBernSwitzerland
- Department for BioMedical Research (DBMR)University of BernBernSwitzerland
- University Children's Hospital Basel (UKBB)BaselSwitzerland
- Division of Respiratory MedicineUniversity Children's Hospital ZurichZurichSwitzerland
| | - Marco P. Alves
- Institute of Virology and ImmunologyBernSwitzerland
- Department of Infectious Diseases and PathobiologyVetsuisse FacultyUniversity of BernBernSwitzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and AllergologyDepartment of Paediatrics, InselspitalBern University HospitalUniversity of BernBernSwitzerland
- Department for BioMedical Research (DBMR)University of BernBernSwitzerland
- University Children's Hospital Basel (UKBB)BaselSwitzerland
| |
Collapse
|
25
|
Primavessy D, Metz J, Schnur S, Schneider M, Lehr CM, Hittinger M. Pulmonary in vitro instruments for the replacement of animal experiments. Eur J Pharm Biopharm 2021; 168:62-75. [PMID: 34438019 DOI: 10.1016/j.ejpb.2021.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/19/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022]
Abstract
Advanced in vitro systems often combine a mechanical-physical instrument with a biological component e.g. cell culture models. For testing of aerosols, it is of advantage to consider aerosol behavior, particle deposition and lung region specific cell lines. Although there are many good reviews on the selection of cell cultures, articles on instruments are rare. This article focuses on the development of in vitro instruments targeting the exposure of aerosols on cell cultures. In this context, guidelines for toxicity investigation are taken into account as the aim of new methods must be the prediction of human relevant data and the replacement of existing animal experiments. We provide an overview on development history of research-based instruments from a pharmaceutical point of view. The standardized commercial devices resulting from the research-based instruments are presented and the future perspectives on pulmonary in vitro devices are discussed.
Collapse
Affiliation(s)
- Daniel Primavessy
- Department of Drug Delivery, PharmBioTec Research and Development GmbH, Saarbrücken, Germany.
| | - Julia Metz
- Department of Drug Delivery, PharmBioTec Research and Development GmbH, Saarbrücken, Germany
| | - Sabrina Schnur
- Department of Drug Delivery, PharmBioTec Research and Development GmbH, Saarbrücken, Germany; Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, PharmBioTec Research and Development GmbH, Saarbrücken, Germany; Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Marius Hittinger
- Department of Drug Delivery, PharmBioTec Research and Development GmbH, Saarbrücken, Germany; 3RProducts Marius Hittinger, Blieskastel, Germany
| |
Collapse
|
26
|
Faber SC, McNabb NA, Ariel P, Aungst ER, McCullough SD. Exposure Effects Beyond the Epithelial Barrier: Transepithelial Induction of Oxidative Stress by Diesel Exhaust Particulates in Lung Fibroblasts in an Organotypic Human Airway Model. Toxicol Sci 2021; 177:140-155. [PMID: 32525552 DOI: 10.1093/toxsci/kfaa085] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In vitro bronchial epithelial monoculture models have been pivotal in defining the adverse effects of inhaled toxicant exposures; however, they are only representative of one cellular compartment and may not accurately reflect the effects of exposures on other cell types. Lung fibroblasts exist immediately beneath the bronchial epithelial barrier and play a central role in lung structure and function, as well as disease development and progression. We tested the hypothesis that in vitro exposure of a human bronchial epithelial cell barrier to the model oxidant diesel exhaust particulates caused transepithelial oxidative stress in the underlying lung fibroblasts using a human bronchial epithelial cell and lung fibroblast coculture model. We observed that diesel exhaust particulates caused transepithelial oxidative stress in underlying lung fibroblasts as indicated by intracellular accumulation of the reactive oxygen species hydrogen peroxide, oxidation of the cellular antioxidant glutathione, activation of NRF2, and induction of oxidative stress-responsive genes. Further, targeted antioxidant treatment of lung fibroblasts partially mitigated the oxidative stress response gene expression in adjacent human bronchial epithelial cells during diesel exhaust particulate exposure. This indicates that exposure-induced oxidative stress in the airway extends beyond the bronchial epithelial barrier and that lung fibroblasts are both a target and a mediator of the adverse effects of inhaled chemical exposures despite being separated from the inhaled material by an epithelial barrier. These findings illustrate the value of coculture models and suggest that transepithelial exposure effects should be considered in inhalation toxicology research and testing.
Collapse
Affiliation(s)
- Samantha C Faber
- Curriculum in Toxicology and Environmental Medicine, UNC Chapel Hill, Chapel Hill, North Carolina 27599
| | - Nicole A McNabb
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, North Carolina 27599
| | - Pablo Ariel
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Emily R Aungst
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, North Carolina 27599
| | - Shaun D McCullough
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Chapel Hill, North Carolina 27599
| |
Collapse
|
27
|
Abstract
Since the industrial revolution, air pollution has become a major problem causing several health problems involving the airways as well as the cardiovascular, reproductive, or neurological system. According to the WHO, about 3.6 million deaths every year are related to inhalation of polluted air, specifically due to pulmonary diseases. Polluted air first encounters the airways, which are a major human defense mechanism to reduce the risk of this aggressor. Air pollution consists of a mixture of potentially harmful compounds such as particulate matter, ozone, carbon monoxide, volatile organic compounds, and heavy metals, each having its own effects on the human body. In the last decades, a lot of research investigating the underlying risks and effects of air pollution and/or its specific compounds on the airways, has been performed, involving both in vivo and in vitro experiments. The goal of this review is to give an overview of the recent data on the effects of air pollution on healthy and diseased airways or models of airway disease, such as asthma or chronic obstructive pulmonary disease. Therefore, we focused on studies involving pollution and airway symptoms and/or damage both in mice and humans.
Collapse
|
28
|
Song N, Wang W, Wang Y, Guan Y, Xu S, Guo MY. Hydrogen sulfide of air induces macrophage extracellular traps to aggravate inflammatory injury via the regulation of miR-15b-5p on MAPK and insulin signals in trachea of chickens. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 771:145407. [PMID: 33548704 DOI: 10.1016/j.scitotenv.2021.145407] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
Hydrogen sulfide (H2S) is an environmental contaminant to cause the airway damage. The release of macrophage extracellular traps (METs) is the mechanism of immune protection to harmful stimulation via microRNAs, but excessive METs cause the injury. However, few studies have attempted to interpret the mechanism of an organism injury due to H2S via METs in chickens. Here, we investigated the transcriptome profiles, pathological morphologic changes and METs release from chicken trachea after H2S exposure. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that 10 differentially expressed genes were related to the METs release, the MAPK and insulin signaling pathways. Morphological and immunofluorescence analysis showed that H2S caused airway injury and MET release. H2S activated the targeting effect of miRNA-15b-5p on activating transcription factor 2 (ATF2). Western blotting and real time quantitative PCR results showed that H2S down-regulated the levels of dual specificity protein phosophatase1 (DUSP1) but up-regulated p38 MAP Kinase (p38) in the MAPK signal pathway. And the expression of phosphoinositide-dependent protein kinase 1 (PDK1), serine/threonine kinase (Akt), and protein kinase ζ subtypes (PKCζ) in the insulin signal pathway were increased after H2S exposure. These promoted the release of myeloperoxidase (MPO) and degradation histone 4 (H4) to induce the release of METs. Taken together, miR-15b-5p targeted ATF2 to mediate METs release, which triggered trachea inflammatory injury via MAPK and insulin signals after H2S exposure. These results will provide new insights into the toxicological mechanisms of H2S and environmental ecotoxicology.
Collapse
Affiliation(s)
- Nuan Song
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Yalin Guan
- College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China
| | - Meng-Yao Guo
- College of Veterinary Medicine, Northeast Agricultural University; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin 150030, China.
| |
Collapse
|
29
|
Nossa R, Costa J, Cacopardo L, Ahluwalia A. Breathing in vitro: Designs and applications of engineered lung models. J Tissue Eng 2021; 12:20417314211008696. [PMID: 33996022 PMCID: PMC8107677 DOI: 10.1177/20417314211008696] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review is to provide a systematic design guideline to users, particularly engineers interested in developing and deploying lung models, and biologists seeking to identify a suitable platform for conducting in vitro experiments involving pulmonary cells or tissues. We first discuss the state of the art on lung in vitro models, describing the most simplistic and traditional ones. Then, we analyze in further detail the more complex dynamic engineered systems that either provide mechanical cues, or allow for more predictive exposure studies, or in some cases even both. This is followed by a dedicated section on microchips of the lung. Lastly, we present a critical discussion of the different characteristics of each type of system and the criteria which may help researchers select the most appropriate technology according to their specific requirements. Readers are encouraged to refer to the tables accompanying the different sections where comprehensive and quantitative information on the operating parameters and performance of the different systems reported in the literature is provided.
Collapse
|
30
|
Cai J, Zang X, Wu Z, Liu J, Wang D. Altered protein S-glutathionylation depicts redox imbalance triggered by transition metal oxide nanoparticles in a breastfeeding system. NANOIMPACT 2021; 22:100305. [PMID: 35559962 DOI: 10.1016/j.impact.2021.100305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/25/2021] [Accepted: 02/19/2021] [Indexed: 06/15/2023]
Abstract
Nanosafety has become a public concern following nanotechnology development. By now, attention has seldom been paid to breastfeeding system, which is constructed by mammary physiological structure and derived substances (endogenous or exogenous), cells, tissues, organs, and individuals (mother and child), connecting environment and organism, and spans across mother-child dyad. Thus, breastfeeding system is a center of nutrients transport and a unique window of toxic susceptibility in the mother-child dyad. We applied metabolomics combined with redox proteomics to depict how nanoparticles cause metabolic burden via their spontaneous redox cycling in lactating mammary glands. Two widely used nanoparticles [titanium dioxide (nTiO2) and zinc oxide (nZnO)] were exposed to lactating mice via intranasal administration. Biodistribution and biopersistence of nTiO2 and nZnO in mammary glands destroyed its structure, reflective of significantly reduced claudin-3 protein level by 32.1% (P < 0.01) and 47.8% (P < 0.01), and significantly increased apoptosis index by 85.7 (P < 0.01) and 100.3 (P < 0.01) fold change, respectively. Airway exposure of nTiO2 trended to reduced milk production by 22.7% (P = 0.06), while nZnO significantly reduced milk production by 33.0% (P < 0.01). Metabolomics analysis revealed a metabolic shift by nTiO2 or nZnO, such as increased glycolysis (nTiO2: fold enrichment = 3.31, P < 0.05; nZnO: fold enrichment = 3.68, P < 0.05), glutathione metabolism (nTiO2: fold enrichment = 5.57, P < 0.01; nZnO: fold enrichment = 4.43, P < 0.05), and fatty acid biosynthesis (nTiO2: fold enrichment = 3.52, P < 0.05; nZnO: fold enrichment = 3.51, P < 0.05) for tissue repair at expense of lower milk fat synthesis (35.7% reduction by nTiO2; 51.8% reduction by nZnO), and finally led to oxidative stress of mammary glands. The increased GSSG/GSH ratio (57.5% increase by nTiO2; 105% increase by nZnO) with nanoparticle exposure confirmed an alteration in the redox state and a metabolic shift in mammary glands. Redox proteomics showed that nanoparticles induced S-glutathionylation (SSG) modification at Cys sites of proteins in a nanoparticle type-dependent manner. The nTiO2 induced more protein SSG modification sites (nTiO2: 21; nZnO:16), whereas nZnO induced fewer protein SSG modification sites but at deeper SSG levels (26.6% higher in average of nZnO than that of nTiO2). In detail, SSG modification by nTiO2 was characterized by Ltf at Cys423 (25.3% increase), and Trf at Cys386;395;583 (42.3%, 42.3%, 22.8% increase) compared with control group. While, SSG modification by nZnO was characterized by Trfc at Cys365 (71.3% increase) and Fasn at Cys1010 (41.0% increase). The discovery of SSG-modified proteins under airway nanoparticle exposure further supplemented the oxidative stress index and mammary injury index, and deciphered precise mechanisms of nanotoxicity into a molecular level. The unique quantitative site-specific redox proteomics and metabolomics can serve as a new technique to identify nanotoxicity and provide deep insights into nanoparticle-triggered oxidative stress, contributing to a healthy breastfeeding environment.
Collapse
Affiliation(s)
- Jie Cai
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Xinwei Zang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Zezhong Wu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China
| | - Jianxin Liu
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| | - Diming Wang
- College of Animal Sciences, Dairy Science Institute, MOE Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou 310029, PR China.
| |
Collapse
|
31
|
He RW, Braakhuis HM, Vandebriel RJ, Staal YC, Gremmer ER, Fokkens PH, Kemp C, Vermeulen J, Westerink RH, Cassee FR. Optimization of an air-liquid interface in vitro cell co-culture model to estimate the hazard of aerosol exposures. JOURNAL OF AEROSOL SCIENCE 2021; 153:105703. [PMID: 33658726 PMCID: PMC7874005 DOI: 10.1016/j.jaerosci.2020.105703] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Inhalation exposure to environmental and occupational aerosol contaminants is associated with many respiratory health problems. To realistically mimic long-term inhalation exposure for toxicity testing, lung epithelial cells need to maintained and exposed under air-liquid interface (ALI) conditions for a prolonged period of time. In addition, to study cellular responses to aerosol particles, lung epithelial cells have to be co-cultured with macrophages. To that aim, we evaluated human bronchial epithelial Calu-3, 16HBE14o- (16HBE), H292, and BEAS-2B cell lines with respect to epithelial morphology, barrier function and cell viability under prolonged ALI culture conditions. Only the Calu-3 cells can retain the monolayer structure and maintain a strong tight junction under long-term ALI culture at least up to 2 weeks. As such, Calu-3 cells were applied as the structural barrier to create co-culture models with human monocyte-derived macrophages (MDMs) and THP-1 derived macrophages (TDMs). Adhesion of macrophages onto the epithelial monolayer was allowed for 4 h with a density of 5 × 104 macrophages/cm2. In comparison to the Calu-3 mono-culture model, Calu-3 + TDM and Calu-3 + MDM co-culture models showed an increased sensitivity in inflammatory responses to lipopolysaccharide (LPS) aerosol at Day 1 of co-culture, with the Calu-3 + MDM model giving a stronger response than Calu-3 + TDM. Therefore, the epithelial monolayer integrity and increased sensitivity make the Calu-3 + MDM co-culture model a preferred option for ALI exposure to inhaled aerosols for toxicity testing.
Collapse
Affiliation(s)
- Rui-Wen He
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80178, 3508, TD, Utrecht, the Netherlands
| | - Hedwig M. Braakhuis
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Rob J. Vandebriel
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Yvonne C.M. Staal
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Eric R. Gremmer
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Paul H.B. Fokkens
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Claudia Kemp
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Jolanda Vermeulen
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
| | - Remco H.S. Westerink
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80178, 3508, TD, Utrecht, the Netherlands
| | - Flemming R. Cassee
- National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands
- Institute for Risk Assessment Sciences, Utrecht University, P.O. Box 80178, 3508, TD, Utrecht, the Netherlands
- Corresponding author. National Institute for Public Health and the Environment (RIVM), P.O. Box 1, 3720, BA, Bilthoven, the Netherlands.
| |
Collapse
|
32
|
Daniel S, Phillippi D, Schneider LJ, Nguyen KN, Mirpuri J, Lund AK. Exposure to diesel exhaust particles results in altered lung microbial profiles, associated with increased reactive oxygen species/reactive nitrogen species and inflammation, in C57Bl/6 wildtype mice on a high-fat diet. Part Fibre Toxicol 2021; 18:3. [PMID: 33419468 PMCID: PMC7796587 DOI: 10.1186/s12989-020-00393-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Exposure to traffic-generated emissions is associated with the development and exacerbation of inflammatory lung disorders such as chronic obstructive pulmonary disorder (COPD) and idiopathic pulmonary fibrosis (IPF). Although many lung diseases show an expansion of Proteobacteria, the role of traffic-generated particulate matter pollutants on the lung microbiota has not been well-characterized. Thus, we investigated the hypothesis that exposure to diesel exhaust particles (DEP) can alter commensal lung microbiota, thereby promoting alterations in the lung's immune and inflammatory responses. We aimed to understand whether diet might also contribute to the alteration of the commensal lung microbiome, either alone or related to exposure. To do this, we used male C57Bl/6 mice (4-6-week-old) on either regular chow (LF) or high-fat (HF) diet (45% kcal fat), randomly assigned to be exposed via oropharyngeal aspiration to 35 μg DEP, suspended in 35 μl 0.9% sterile saline or sterile saline only (control) twice a week for 30 days. A separate group of study animals on the HF diet was concurrently treated with 0.3 g/day of Winclove Ecologic® Barrier probiotics in their drinking water throughout the study. RESULTS Our results show that DEP-exposure increases lung tumor necrosis factor (TNF)-α, interleukin (IL)-10, Toll-like receptor (TLR)-2, TLR-4, and the nuclear factor kappa B (NF-κB) histologically and by RT-qPCR, as well as Immunoglobulin A (IgA) and Immunoglobulin G (IgG) in the bronchoalveolar lavage fluid (BALF), as quantified by ELISA. We also observed an increase in macrophage infiltration and peroxynitrite, a marker of reactive oxygen species (ROS) + reactive nitrogen species (RNS), immunofluorescence staining in the lungs of DEP-exposed and HF-diet animals, which was further exacerbated by concurrent DEP-exposure and HF-diet consumption. Histological examinations revealed enhanced inflammation and collagen deposition in the lungs DEP-exposed mice, regardless of diet. We observed an expansion of Proteobacteria, by qPCR of bacterial 16S rRNA, in the BALF of DEP-exposed mice on the HF diet, which was diminished with probiotic-treatment. CONCLUSIONS Our findings suggest that exposure to DEP causes persistent and sustained inflammation and bacterial alterations in a ROS-RNS mediated fashion, which is exacerbated by concurrent consumption of an HF diet.
Collapse
Affiliation(s)
- Sarah Daniel
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Leah J Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Kayla N Nguyen
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA
| | - Julie Mirpuri
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Amie K Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, EESAT - 215, 1704 W. Mulberry, Denton, TX, 76201, USA.
| |
Collapse
|
33
|
Addressing the challenges of E-cigarette safety profiling by assessment of pulmonary toxicological response in bronchial and alveolar mucosa models. Sci Rep 2020; 10:20460. [PMID: 33235237 PMCID: PMC7686373 DOI: 10.1038/s41598-020-77452-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
Limited toxicity data on electronic cigarette (ECIG) impede evidence-based policy recommendations. We compared two popular mixed fruit flavored ECIG-liquids with and without nicotine aerosolized at 40 W (E-smoke) with respect to particle number concentrations, chemical composition, and response on physiologically relevant human bronchial and alveolar lung mucosa models cultured at air–liquid interface. E-smoke was characterized by significantly increased particle number concentrations with increased wattage (25, 40, and 55 W) and nicotine presence. The chemical composition of E-smoke differed across the two tested flavors in terms of cytotoxic compounds including p-benzoquinone, nicotyrine, and flavoring agents (for example vanillin, ethyl vanillin). Significant differences in the expression of markers for pro-inflammation, oxidative stress, tissue injury/repair, alarm anti-protease, anti-microbial defense, epithelial barrier function, and epigenetic modification were observed between the flavors, nicotine content, and/ or lung models (bronchial or alveolar). Our findings indicate that ECIG toxicity is influenced by combination of multiple factors including flavor, nicotine content, vaping regime, and the region of respiratory tree (bronchial or alveolar). Toxic chemicals and flavoring agents detected in high concentrations in the E-smoke of each flavor warrant independent evaluation for their specific role in imparting toxicity. Therefore, multi-disciplinary approaches are warranted for comprehensive safety profiling of ECIG.
Collapse
|
34
|
Smyth T, Veazey J, Eliseeva S, Chalupa D, Elder A, Georas SN. Diesel exhaust particle exposure reduces expression of the epithelial tight junction protein Tricellulin. Part Fibre Toxicol 2020; 17:52. [PMID: 33059747 PMCID: PMC7560077 DOI: 10.1186/s12989-020-00383-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND While exposure to diesel exhaust particles has been linked to aberrant immune responses in allergic diseases such as asthma, little attention has been paid to their effects on the airway epithelial barrier. In this study, we sought to determine the effect of diesel exhaust exposure on airway epithelial barrier function and composition using in vitro and in vivo model systems. METHODS 16HBE14o- human bronchial epithelial cells were grown on collagen coated Transwell inserts and exposed to 5 to 50 μg/cm2 SRM 2975 diesel particulate matter (DEP) suspended in cell culture medium or vehicle controls. Changes in barrier function were assessed by measuring transepithelial electrical resistance (TEER) and permeability to 4 kDa FITC Dextran. Neonatal BALB/c mice were exposed to aerosolized DEP (255 ± 89 μg/m3; 2 h per day for 5 days) and changes in the tight junction protein Tricellulin were assessed 2 weeks post exposure. RESULTS A six-hour incubation of epithelial cells with diesel exhaust particles caused a significant concentration-dependent reduction in epithelial barrier integrity as measured by decreased TEER and increased permeability to 4 kDa FITC-Dextran. This reduction in epithelial barrier integrity corresponded to a significant reduction in expression of the tight junction protein Tricellulin. siRNA mediated knockdown of Tricellulin recapitulated changes in barrier function caused by DEP exposure. Neonatal exposure to aerosolized DEP caused a significant reduction in lung Tricellulin 2 weeks post exposure at both the protein and mRNA level. CONCLUSION Short term exposure to DEP causes a significant reduction in epithelial barrier integrity through a reduction in the tight junction protein Tricellulin. Neonatal exposure to aerosolized DEP caused a significant and sustained reduction in Tricellulin protein and mRNA in the lung, suggesting that early life exposure to inhaled DEP may cause lasting changes in airway epithelial barrier function.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Janelle Veazey
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA
| | - Sophia Eliseeva
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Box 692, 601 Elmwood Ave, University of Rochester, Rochester, NY, 14627, USA
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Alison Elder
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, USA.
- Department of Medicine, Pulmonary and Critical Care, University of Rochester, Box 692, 601 Elmwood Ave, University of Rochester, Rochester, NY, 14627, USA.
| |
Collapse
|
35
|
Clinical, Epidemiological and Experimental Approaches to Assess Adverse Health Outcomes of Indoor Biomass Smoke Exposure: Conclusions from An Indo-Swedish Workshop in Mysuru, January 2020. TOXICS 2020; 8:toxics8030068. [PMID: 32899560 PMCID: PMC7560295 DOI: 10.3390/toxics8030068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022]
Abstract
This report summarizes the outcome of a workshop held in Mysuru, India in January 2020 addressing the adverse health effects of exposure to biomass smoke (BMS). The aim of the workshop was to identify uncertainties and gaps in knowledge and possible methods to address them in the Mysuru study on Determinants of Health in Rural Adults (MUDHRA) cohort. Specific aims were to discuss the possibility to improve and introduce new screening methods for exposure and effect, logistic limitations and other potential obstacles, and plausible strategies to overcome these in future studies. Field visits were included in the workshop prior to discussing these issues. The workshop concluded that multi-disciplinary approaches to perform: (a) indoor and personalized exposure assessment; (b) clinical and epidemiological field studies among children, adolescents, and adults; (c) controlled exposure experiments using physiologically relevant in vitro and in vivo models to understand molecular patho-mechanisms are warranted to dissect BMS-induced adverse health effects. It was perceived that assessment of dietary exposure (like phytochemical index) may serve as an important indicator for understanding potential protective mechanisms. Well trained field teams and close collaboration with the participating hospital were identified as the key requirements to successfully carry out the study objectives.
Collapse
|
36
|
Zhang X, Zhang Y, Meng Q, Sun H, Wu S, Xu J, Yun J, Yang X, Li B, Zhu H, Xue L, Li X, Chen R. MicroRNA-382-5p is involved in pulmonary inflammation induced by fine particulate matter exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 262:114278. [PMID: 32146367 DOI: 10.1016/j.envpol.2020.114278] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/31/2020] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
Exposure to atmospheric particulate matter (PM) has been related to the increasing incidence and mortality of pulmonary diseases, where microRNAs (miRNAs) play significant roles in these biological and pathological processes. In the present study, we found that miR-382-5p played an anti-inflammatory role in pulmonary inflammation induced by fine particulate matter (PM2.5) or diesel exhaust particles (DEPs) in vitro and in vivo. The expression level of miR-382-5p was downregulated, while its target gene, namely CXCL12, was elevated in HBE cells after exposure to PM2.5 or DEPs. Mechanistically, PM2.5 or DEPs exposure increased CXCL12/MMP9 expression via miR-382-5p inhibition, subsequently triggered pulmonary inflammation. Furthermore, antagonizing the function of CXCL12 significantly reduced the expression of MMP9 and local inflammation induced by PM2.5 or DEPs. PM2.5 or DEPs caused apoptosis and G1 phase arrest could be partially restored by overexpression of miR-382-5p and antagonism of CXCL12. In a murine model, enhanced miR-382-5p expression effectively reduced expression levels of CXCL12, MMP9 and inflammatory cytokines, hereby protected lung tissues against PM2.5 or DEPs-induced lesions. Collectively, the miR-382-5p/CXCL12/MMP9 pathway may provide a mechanism, which mediates inflammatory response to PM2.5 or DEPs exposure.
Collapse
Affiliation(s)
- Xinwei Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, Hebei Province, China
| | - Qingtao Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Hao Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Shenshen Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jie Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Jun Yun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Xi Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Hao Zhu
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, Hebei Province, China
| | - Ling Xue
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, Hebei Province, China
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
37
|
Nanodomains in cardiopulmonary disorders and the impact of air pollution. Biochem Soc Trans 2020; 48:799-811. [PMID: 32597478 PMCID: PMC7329344 DOI: 10.1042/bst20190250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Air pollution is a major environmental threat and each year about 7 million people reported to die as a result of air pollution. Consequently, exposure to air pollution is linked to increased morbidity and mortality world-wide. Diesel automotive engines are a major source of urban air pollution in the western societies encompassing particulate matter and diesel exhaust particles (DEP). Air pollution is envisioned as primary cause for cardiovascular dysfunction, such as ischemic heart disease, cardiac dysrhythmias, heart failure, cerebrovascular disease and stroke. Air pollution also causes lung dysfunction, such as chronic obstructive pulmonary disease (COPD), asthma, idiopathic pulmonary fibrosis (IPF), and specifically exacerbations of these diseases. DEP induces inflammation and reactive oxygen species production ultimately leading to mitochondrial dysfunction. DEP impair structural cell function and initiate the epithelial-to-mesenchymal transition, a process leading to dysfunction in endothelial as well as epithelial barrier, hamper tissue repair and eventually leading to fibrosis. Targeting cyclic adenosine monophosphate (cAMP) has been implicated to alleviate cardiopulmonary dysfunction, even more intriguingly cAMP seems to emerge as a potent regulator of mitochondrial metabolism. We propose that targeting of the mitochondrial cAMP nanodomain bear the therapeutic potential to diminish air pollutant — particularly DEP — induced decline in cardiopulmonary function.
Collapse
|
38
|
Ehrmann S, Schmid O, Darquenne C, Rothen-Rutishauser B, Sznitman J, Yang L, Barosova H, Vecellio L, Mitchell J, Heuze-Vourc’h N. Innovative preclinical models for pulmonary drug delivery research. Expert Opin Drug Deliv 2020; 17:463-478. [PMID: 32057260 PMCID: PMC8083945 DOI: 10.1080/17425247.2020.1730807] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/11/2020] [Indexed: 02/08/2023]
Abstract
Introduction: Pulmonary drug delivery is a complex field of research combining physics which drive aerosol transport and deposition and biology which underpins efficacy and toxicity of inhaled drugs. A myriad of preclinical methods, ranging from in-silico to in-vitro, ex-vivo and in-vivo, can be implemented.Areas covered: The present review covers in-silico mathematical and computational fluid dynamics modelization of aerosol deposition, cascade impactor technology to estimated drug delivery and deposition, advanced in-vitro cell culture methods and associated aerosol exposure, lung-on-chip technology, ex-vivo modeling, in-vivo inhaled drug delivery, lung imaging, and longitudinal pharmacokinetic analysis.Expert opinion: No single preclinical model can be advocated; all methods are fundamentally complementary and should be implemented based on benefits and drawbacks to answer specific scientific questions. The overall best scientific strategy depends, among others, on the product under investigations, inhalation device design, disease of interest, clinical patient population, previous knowledge. Preclinical testing is not to be separated from clinical evaluation, as small proof-of-concept clinical studies or conversely large-scale clinical big data may inform preclinical testing. The extend of expertise required for such translational research is unlikely to be found in one single laboratory calling for the setup of multinational large-scale research consortiums.
Collapse
Affiliation(s)
- Stephan Ehrmann
- CHRU Tours, Médecine Intensive Réanimation, CIC INSERM 1415, CRICS-TriggerSep network, Tours France
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München – German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Chantal Darquenne
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC0623A, La Jolla, CA 92093-0623, United States
| | | | - Josue Sznitman
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Julius Silver building, Office 246, Haifa 32000, Israel
| | - Lin Yang
- Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München – German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Hana Barosova
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, Switzerland
| | - Laurent Vecellio
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| | - Jolyon Mitchell
- Jolyon Mitchell Inhaler Consulting Services Inc., 1154 St. Anthony Road, London, Ontario, Canada, N6H 2R1
| | - Nathalie Heuze-Vourc’h
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
39
|
Cappellini F, Di Bucchianico S, Karri V, Latvala S, Malmlöf M, Kippler M, Elihn K, Hedberg J, Odnevall Wallinder I, Gerde P, Karlsson HL. Dry Generation of CeO 2 Nanoparticles and Deposition onto a Co-Culture of A549 and THP-1 Cells in Air-Liquid Interface-Dosimetry Considerations and Comparison to Submerged Exposure. NANOMATERIALS 2020; 10:nano10040618. [PMID: 32230801 PMCID: PMC7221976 DOI: 10.3390/nano10040618] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/09/2020] [Accepted: 03/24/2020] [Indexed: 02/04/2023]
Abstract
Relevant in vitro assays that can simulate exposure to nanoparticles (NPs) via inhalation are urgently needed. Presently, the most common method employed is to expose lung cells under submerged conditions, but the cellular responses to NPs under such conditions might differ from those observed at the more physiological air-liquid interface (ALI). The aim of this study was to investigate the cytotoxic and inflammatory potential of CeO2 NPs (NM-212) in a co-culture of A549 lung epithelial cells and differentiated THP-1 cells in both ALI and submerged conditions. Cellular dose was examined quantitatively using inductively coupled plasma mass spectrometry (ICP-MS). The role of serum and LPS-priming for IL-1β release was further tested in THP-1 cells in submerged exposure. An aerosol of CeO2 NPs was generated by using the PreciseInhale® system, and NPs were deposited on the co-culture using XposeALI®. No or minor cytotoxicity and no increased release of inflammatory cytokines (IL-1β, IL-6, TNFα, MCP-1) were observed after exposure of the co-culture in ALI (max 5 µg/cm2) or submerged (max 22 µg/cm2) conditions. In contrast, CeO2 NPs cause clear IL-1β release in monocultures of macrophage-like THP-1, independent of the presence of serum and LPS-priming. This study demonstrates a useful approach for comparing effects at various in-vitro conditions.
Collapse
Affiliation(s)
- Francesca Cappellini
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
| | - Sebastiano Di Bucchianico
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
- Comprehensive Molecular Analytics, Helmholtz Zentrum München, 81379 München, Germany
| | - Venkatanaidu Karri
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
| | - Siiri Latvala
- Department of Environmental Science, Stockholm University, Stockholm11418, Sweden
| | - Maria Malmlöf
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
- Inhalation Sciences, Hälsovägen 7-9, 141 57 Huddinge, Sweden
| | - Maria Kippler
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
| | - Karine Elihn
- Department of Environmental Science, Stockholm University, Stockholm11418, Sweden
| | - Jonas Hedberg
- KTH Royal Institute of Technology, Department of Chemistry, Division of Surface and Corrosion Science, 114 28 Stockholm, Sweden
| | - Inger Odnevall Wallinder
- KTH Royal Institute of Technology, Department of Chemistry, Division of Surface and Corrosion Science, 114 28 Stockholm, Sweden
| | - Per Gerde
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
- Inhalation Sciences, Hälsovägen 7-9, 141 57 Huddinge, Sweden
| | - Hanna L. Karlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17177 Sweden
- Correspondence:
| |
Collapse
|
40
|
Li X, Zhang Y, Li B, Yang H, Cui J, Li X, Zhang X, Sun H, Meng Q, Wu S, Li S, Wang J, Aschner M, Chen R. Activation of NLRP3 in microglia exacerbates diesel exhaust particles-induced impairment in learning and memory in mice. ENVIRONMENT INTERNATIONAL 2020; 136:105487. [PMID: 31999974 DOI: 10.1016/j.envint.2020.105487] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/19/2019] [Accepted: 01/11/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND The major components of traffic pollution particulate matter, diesel exhaust particles (DEPs), are airborne ultrafine particles (UFPs). DEPs can enter the central nervous system (CNS), where they may cause neurotoxicity. METHODS We established murine models with intranasal DEPs instillation in male C57BL/6 and Nlrp3 knock-out (Nlrp3-/-) mice to investigate the effects of DEPs exposure on murine neurobehaviors and related mechanisms. Morris water maze (MWM) tests were performed to evaluate the learning and memory behaviors of mice following DEPs instillation. Metabolomics were assessed using an gas chromatography system coupled to a mass spectrometer. Real-time PCR and immunohistochemistry assays were used to analyze the mRNA and protein expression levels of target genes. Murine microglia, BV2 cells were employed to assay the effects of DEPs exposure in vitro. RESULTS Intranasal administration of DEPs in mice led to impairment in hippocampal-dependent learning and memory. Moreover, this phenotype was linked to increased number of Iba-1+ microglia and NLRP3 inflammasome, as well as suppression of mitochondrial gene expression in the hippocampus of mice exposed to DEPs. Nlrp3-/- mice were resistant to DEPs-induced learning and memory impairment, concomitant with protection against the suppression of mitochondrial gene expression. Murine microglia cells (BV2) were exposed to DEPs in vitro and taurine was identified as one of the significantly suppressed metabolites in DEPs-treated microglia by metabolomics analysis. Supplementation with taurine efficiently rescued learning, memory and mitochondrial gene expression levels in the hippocampus of DEPs-exposed mice. CONCLUSIONS Mechanistically, our study revealed that microglia-mediated NLRP3 inflammasome activation plays a deleterious role in DEPs-induced neurotoxicity by inhibiting mitochondrial gene expression. These results shed novel light on the potential value of nutritional supplementation against DEPs-induced neurotoxicity in individuals exposed to severe airborne traffic-related air pollutions.
Collapse
Affiliation(s)
- Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yanshu Zhang
- Laboratory Animal Center, North China University of Science and Technology, Tangshan 063210, China
| | - Bin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Hongbao Yang
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jian Cui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xiaoyan Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xinwei Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Hao Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Qingtao Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shenshen Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shuang Li
- Laboratory Animal Center, North China University of Science and Technology, Tangshan 063210, China
| | - Jianbo Wang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
41
|
Flies EJ, Mavoa S, Zosky GR, Mantzioris E, Williams C, Eri R, Brook BW, Buettel JC. Urban-associated diseases: Candidate diseases, environmental risk factors, and a path forward. ENVIRONMENT INTERNATIONAL 2019; 133:105187. [PMID: 31648161 DOI: 10.1016/j.envint.2019.105187] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 05/24/2023]
Abstract
BACKGROUND Cities are home to over half the global population; that proportion is expected to rise to 70% by mid-century. The urban environment differs greatly from that in which humans evolved, with potentially important consequences for health. Rates for allergic, inflammatory and auto-immune diseases appear to rise with urbanization and be higher in the more urbanized nations of the world which has led some to suggest that cities promote the occurrence of these diseases. However, there are no syntheses outlining what urban-associated diseases are and what characteristics of cities promote their occurrence. OBJECTIVES To synthesize the current understanding of "urban-associated diseases", and discover the common, potentially modifiable features of cities that may be driving these associations. METHODS We focus on any diseases that have been associated with cities or are particularly prominent in today's urban societies. We draw on expertise across diverse health fields to examine the evidence for urban connections and drivers. DISCUSSION We found evidence for urban associations across allergic, auto-immune, inflammatory, lifestyle and infectious disease categories. Some conditions (e.g. obesity and diabetes) have complex relationships with cities that have been insufficiently explored. Other conditions (e.g. allergies and asthma) have more evidence demonstrating their relationship with cities and the mechanisms driving that association. Unsurprisingly, air pollution was the characteristic of cities most frequently associated with disease. Other identified urban risk factors are not as widely known: altered microbial exposure and a disconnect from environmental microbiomes, vitamin D deficiency, noise and light pollution, and a transient, over-crowded, impoverished population. However, many complexities and caveats to these relationships beg clarification; we highlight the current knowledge gaps and outline ways to fill those gaps. Identifying urban-associated diseases and their drivers will allow us to prepare for the urban-disease burden of the future and create healthy cities that mitigate that disease burden.
Collapse
Affiliation(s)
- Emily J Flies
- School of Natural Sciences, College of Science and Engineering, University of Tasmania, Hobart, Australia.
| | - Suzanne Mavoa
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Australia; School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Evangeline Mantzioris
- School of Pharmacy and Medical Sciences & Alliance for Research in Exercise, Nutrition and Activity (ARENA), University of South Australia, Adelaide, Australia
| | - Craig Williams
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Barry W Brook
- School of Natural Sciences, College of Science and Engineering, University of Tasmania, Hobart, Australia
| | - Jessie C Buettel
- School of Natural Sciences, College of Science and Engineering, University of Tasmania, Hobart, Australia
| |
Collapse
|
42
|
Gibbs JL, Dallon BW, Lewis JB, Walton CM, Arroyo JA, Reynolds PR, Bikman BT. Diesel Exhaust Particle Exposure Compromises Alveolar Macrophage Mitochondrial Bioenergetics. Int J Mol Sci 2019; 20:ijms20225598. [PMID: 31717476 PMCID: PMC6888061 DOI: 10.3390/ijms20225598] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022] Open
Abstract
Diesel exhaust particles (DEPs) are known pathogenic pollutants that constitute a significant quantity of air pollution. Given the ubiquitous presence of macrophages throughout the body, including the lungs, as well as their critical role in tissue and organismal metabolic function, we sought to determine the effect of DEP exposure on macrophage mitochondrial function. Following daily DEP exposure in mice, pulmonary macrophages were isolated for mitochondrial analyses, revealing reduced respiration rates and dramatically elevated H2O2 levels. Serum ceramides and inflammatory cytokines were increased. To determine the degree to which the changes in mitochondrial function in macrophages were not dependent on any cross-cell communication, primary pulmonary murine macrophages were used to replicate the DEP exposure in a cell culture model. We observed similar changes as seen in pulmonary macrophages, namely diminished mitochondrial respiration, but increased H2O2 production. Interestingly, when treated with myriocin to inhibit ceramide biosynthesis, these DEP-induced mitochondrial changes were mitigated. Altogether, these data suggest that DEP exposure may compromise macrophage mitochondrial and whole-body function via pathologic alterations in macrophage ceramide metabolism.
Collapse
Affiliation(s)
- Jonathan L. Gibbs
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Blake W. Dallon
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Joshua B. Lewis
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Chase M. Walton
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Juan A. Arroyo
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Paul R. Reynolds
- Lung and Placental Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Benjamin T. Bikman
- Metabolism Research Lab, Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
- Correspondence:
| |
Collapse
|
43
|
Shi R, Su WW, Zhu ZT, Guan MY, Cheng KL, Fan WY, Wei GY, Li PB, Yang ZY, Yao HL. Regulation effects of naringin on diesel particulate matter-induced abnormal airway surface liquid secretion. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 63:153004. [PMID: 31301536 DOI: 10.1016/j.phymed.2019.153004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/05/2019] [Accepted: 06/29/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND PM2.5 is closely related to the incidence and mortality of respiratory diseases. Diesel particulate matter (DPM) is the main component of particulate air pollution and an important source of PM2.5. HYPOTHESIS/PURPOSE This study mainly explored the effect of DPM on airway surface liquid (ASL) secretion and the regulation of naringin in this process, to evaluate therapeutic potentials of naringin for the treatment of abnormal secretion of the respiratory tract caused by PM2.5. METHODS The concentration of lysozyme was measured by Lysozyme Assay Kit. Total protein content was determined by the BCA Protein Assay Kit. The concentration of cAMP and MUC5AC, expressions of CFTR, AQP1, and AQP5 proteins were measured by ELISA. Expressions of CFTR, AQP1 and AQP5 mRNA were determined by qPCR. Amount of CFTR on the cell membrane was determined by immunofluorescence. RESULTS The in vitro and in vivo studies had indicated that DPM could inhibit ASL secretion and increased the viscosity of the liquid. Naringin had the functions to attenuate DPM-induced injury, reduce liquid viscosity by reducing MUC5AC and total protein secretion, increase DPM-induced CFTR, AQP1, and AQP5 mRNA and protein expression, positively regulate apical CFTR insertion and promote CFTR activation by increasing intracellular cAMP. CONCLUSION These results demonstrated that naringin had regulating effects on the DPM-induced abnormal secretion of the respiratory tract.
Collapse
Affiliation(s)
- Rui Shi
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei-Wei Su
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Ting Zhu
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Min-Yi Guan
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ke-Ling Cheng
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei-Yang Fan
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Gu-Yi Wei
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pei-Bo Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhong-Yi Yang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hong-Liang Yao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources, Guangdong Engineering & Technology Research Center for Quality and Efficacy Re-evaluation of Post-market Traditional Chinese Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
44
|
Ji J, Ganguly K, Mihai X, Sun J, Malmlöf M, Gerde P, Upadhyay S, Palmberg L. Exposure of normal and chronic bronchitis-like mucosa models to aerosolized carbon nanoparticles: comparison of pro-inflammatory oxidative stress and tissue injury/repair responses. Nanotoxicology 2019; 13:1362-1379. [PMID: 31462114 DOI: 10.1080/17435390.2019.1655600] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Carbon nanoparticles (CNP) are generated by incomplete combustion of diesel engines. Several epidemiological studies associated higher susceptibility to particulate matter related adverse respiratory outcomes with preexisting conditions like chronic bronchitis (CB). Therefore, we compared the effect of CNP exposure on primary bronchial epithelial cells (PBEC) developed in air-liquid interface (ALI) models of normal versus CB-like-mucosa.PBEC cultured at ALI represented normal mucosa (PBEC-ALI). To develop CB-like-mucosa (PBEC-ALI/CB), 1 ng/ml interleukin-13 was added to the basal media of PBEC-ALI culturing. PBEC-ALI and PBEC-ALI/CB were exposed to sham or to aerosolized CNP using XposeALI® system. Protein levels of CXCL-8 and MMP-9 were measured in the basal media using ELISA. Transcript expression of pro-inflammatory (CXCL8, IL6, TNF, NFKB), oxidative stress (HMOX1, SOD3, GSTA1, GPx), tissue injury/repair (MMP9/TIMP1) and bronchial cell type markers (MUC5AC, CC10) were assessed using qRT-PCR.Increased secretion of CXCL-8 and MMP-9 markers was detected 24 h post-exposure in both PBEC-ALI and PBEC-ALI/CB with more pronounced effect in the later. Pro-inflammatory and tissue injury markers were increased at both 6 h and 24 h post-exposure in PBEC-ALI/CB. Oxidative stress markers exhibited similar responses at 6 h and 24 h post-exposure in PBEC-ALI/CB. The club cell specific marker CC10 was increased by 300 fold in PBEC-ALI/CB and 20 fold in PBEC-ALI following CNP exposure.Our data indicates an earlier and stronger reaction of pro-inflammatory, oxidative stress and tissue injury markers in PBEC-ALI/CB models compared to PBEC-ALI models following CNP exposure. The findings may provide insight into the plausible mechanisms of higher susceptibility among predisposed individuals to nanoparticle exposure.
Collapse
Affiliation(s)
- Jie Ji
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Koustav Ganguly
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xenia Mihai
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jitong Sun
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Malmlöf
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Inhalation Sciences Sweden AB, Stockholm, Sweden
| | - Per Gerde
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Inhalation Sciences Sweden AB, Stockholm, Sweden
| | - Swapna Upadhyay
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Palmberg
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Thimraj TA, Sompa SI, Ganguly K, Ernstgård L, Johanson G, Palmberg L, Upadhyay S. Evaluation of diacetyl mediated pulmonary effects in physiologically relevant air-liquid interface models of human primary bronchial epithelial cells. Toxicol In Vitro 2019; 61:104617. [PMID: 31381966 DOI: 10.1016/j.tiv.2019.104617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/03/2019] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
Diacetyl is an artificial flavouring agent, known to cause bronchiolitis obliterans. Diacetyl-induced pulmonary effects were assessed in human primary bronchial epithelial cells (PBEC) cultured at air-liquid interface (ALI). The PBEC-ALI models were exposed to clean air (sham) and diacetyl vapour (1, 3, 10 and 30 ppm) for 30 min. At 6 and 24 h post-exposure, cell medium was sampled for assessment of cytotoxicity measurement, and CXCL8, MMP9 secretion by ELISA. Pro-inflammatory, oxidative stress, tissue injury/repair, anti-protease and beta-defensin markers were assessed using qRT-PCR. Additionally, epidermal growth factor receptor ligands (amphiregulin) and anti-protease (SLPI) were analysed at 6 h, 8 h and 24 h post exposure to 1 and 10 ppm diacetyl. No significant cytotoxicity was observed at any exposure level. MMP9 was significantly increased in both apical and basal media at 24 h. Both SLPI and amphiregulin secretion were significantly increased following exposure to 10 ppm diacetyl. Exposure of PBEC-ALI model to diacetyl vapour resulted in significantly altered transcript expression of pro-inflammatory, oxidative stress, anti-protease, tissue injury/repair markers. Changes in transcript expression of significantly altered markers were more prominent 24 h post-exposure compared to 6 h. This study warrants further mechanistic investigations to elucidate the pulmonary effects of inhaled diacetyl vapour using physiologically relevant in vitro models.
Collapse
Affiliation(s)
- Tania A Thimraj
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Shanzina I Sompa
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Koustav Ganguly
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Lena Ernstgård
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Gunnar Johanson
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Lena Palmberg
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Swapna Upadhyay
- Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
46
|
Kaur K, Jaramillo IC, Mohammadpour R, Sturrock A, Ghandehari H, Reilly C, Paine R, Kelly KE. Effect of collection methods on combustion particle physicochemical properties and their biological response in a human macrophage-like cell line. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2019; 54:1170-1185. [PMID: 31342848 PMCID: PMC6801061 DOI: 10.1080/10934529.2019.1632626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 06/10/2023]
Abstract
In vitro studies are a first step toward understanding the biological effects of combustion-derived particulate matter (cdPM). A vast majority of studies expose cells to cdPM suspensions, which requires a method to collect cdPM and suspend it in an aqueous media. The consequences of different particle collection methods on particle physiochemical properties and resulting biological responses are not fully understood. This study investigated the effect of two common approaches (collection on a filter and a cold plate) and one relatively new (direct bubbling in DI water) approach to particle collection. The three approaches yielded cdPM with differences in particle size distribution, surface area, composition, and oxidative potential. The directly bubbled sample retained the smallest sized particles and the bimodal distribution observed in the gas-phase. The bubbled sample contained ∼50% of its mass as dissolved species and lower molecular weight compounds, not found in the other two samples. These differences in the cdPM properties affected the biological responses in THP-1 cells. The bubbled sample showed greater oxidative potential and cellular reactive oxygen species. The scraped sample induced the greatest TNFα secretion. These findings have implications for in vitro studies of air pollution and for efforts to better understand the underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | - Anne Sturrock
- Division of Pulmonary and Critical Care Medicine, University of Utah
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah
- Department of Bioengineering, University of Utah
| | - Christopher Reilly
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah
- Department of Pharmacology and Toxicology, University of Utah
| | - Robert Paine
- Division of Pulmonary and Critical Care Medicine, University of Utah
| | - Kerry E. Kelly
- Department of Chemical Engineering, University of Utah
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah
| |
Collapse
|
47
|
Kulvinskiene I, Raudoniute J, Bagdonas E, Ciuzas D, Poliakovaite K, Stasiulaitiene I, Zabulyte D, Bironaite D, Rimantas Venskutonis P, Martuzevicius D, Aldonyte R. Lung alveolar tissue destruction and protein citrullination in diesel exhaust-exposed mouse lungs. Basic Clin Pharmacol Toxicol 2019; 125:166-177. [PMID: 30801928 DOI: 10.1111/bcpt.13213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/07/2019] [Indexed: 01/03/2023]
Abstract
Humanity faces an increasing impact of air pollution worldwide, including threats to human health. Air pollutants prompt and promote chronic inflammation, tumourigenesis, autoimmune and other destructive processes in the human body. Post-translational modification of proteins, for example citrullination, results from damaging attacks of pollutants, including smoking, air pollution and others, rendering host tissues immunogenic. Citrullinated proteins and citrullinating enzymes, deiminases, are more prevalent in patients with COPD and correlate with ongoing inflammation and oxidative stress. In this study, we installed an in-house-designed diesel exhaust delivery and cannabidiol vaporization system where mice were exposed to relevant, urban traffic-related levels of diesel exhaust for 14 days and assessed integrity of alveolar tissue, gene expression shifts and changes in protein content in the lungs and other tissues of exposed mice. Systemic presence of modified proteins was also tested. The protective effect of phytocannabinoids was investigated as well. Data obtained in our study show subacute effects of diesel exhaust on mouse lung integrity and protein content. Emphysematous changes are documented in exposed mouse lungs. In parallel, increased levels of citrulline were detected in the alveolar lung tissue and peripheral blood of exposed mice. Pre-treatment with vaporized cannabidiol ameliorated some damaging effects. Results reported hereby provide new insights into subacute lung tissue changes that follow diesel exhaust exposure and suggest possible dietary and/or other therapeutic interventions for maintaining lung health and healthy ageing.
Collapse
Affiliation(s)
- Ieva Kulvinskiene
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Jovile Raudoniute
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Edvardas Bagdonas
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Darius Ciuzas
- Department of Environmental Technologies, Kaunas University of Technology, Kaunas, Lithuania
| | | | - Inga Stasiulaitiene
- Department of Environmental Technologies, Kaunas University of Technology, Kaunas, Lithuania
| | - Danguole Zabulyte
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | - Daiva Bironaite
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| | | | - Dainius Martuzevicius
- Department of Environmental Technologies, Kaunas University of Technology, Kaunas, Lithuania
| | - Ruta Aldonyte
- State Research Institute Center for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|