1
|
Portugal J, Bedia C, Amato F, Juárez-Facio AT, Stamatiou R, Lazou A, Campiglio CE, Elihn K, Piña B. Toxicity of airborne nanoparticles: Facts and challenges. ENVIRONMENT INTERNATIONAL 2024; 190:108889. [PMID: 39042967 DOI: 10.1016/j.envint.2024.108889] [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: 04/18/2024] [Revised: 07/10/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024]
Abstract
Air pollution is one of the most severe environmental healthhazards, and airborne nanoparticles (diameter <100 nm) are considered particularly hazardous to human health. They are produced by various sources such as internal combustion engines, wood and biomass burning, and fuel and natural gas combustion, and their origin, among other parameters, determines their intrinsic toxicity for reasons that are not yet fully understood. Many constituents of the nanoparticles are considered toxic or at least hazardous, including polycyclic aromatic hydrocarbons (PAHs) and heavy metal compounds, in addition to gaseous pollutants present in the aerosol fraction, such as NOx, SO2, and ozone. All these compounds can cause oxidative stress, mitochondrial damage, inflammation in the lungs and other tissues, and cellular organelles. Epidemiological investigations concluded that airborne pollution may affect the respiratory, cardiovascular, and nervous systems. Moreover, particulate matter has been linked to an increased risk of lung cancer, a carcinogenic effect not related to DNA damage, but to the cellular inflammatory response to the pollutants, in which the release of cytokines promotes the proliferation of pre-existing mutated cancer cells. The mechanisms behind toxicity can be investigated experimentally using cell cultures or animal models. Methods for gathering particulate matter have been explored, but standardized protocols are needed to ensure that the samples accurately represent chemical mixtures in the environment. Toxic constituents of nanoparticles can be studied in animal and cellular models, but designing realistic exposure settings is challenging. The air-liquid interface (ALI) system directly exposes cells, mimicking particle inhalation into the lungs. Continuous research and monitoring of nanoparticles and other airborne pollutants is essential for understanding their effects and developing active strategies to mitigate their risks to human and environmental health.
Collapse
Affiliation(s)
- José Portugal
- Institute of Environmental Assessment and Water Research, CSIC, 08034 Barcelona, Spain.
| | - Carmen Bedia
- Institute of Environmental Assessment and Water Research, CSIC, 08034 Barcelona, Spain
| | - Fulvio Amato
- Institute of Environmental Assessment and Water Research, CSIC, 08034 Barcelona, Spain
| | - Ana T Juárez-Facio
- Department of Environmental Science, Stockholm University, 11419 Stockholm, Sweden
| | - Rodopi Stamatiou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Chiara E Campiglio
- Department of Management, Information and Production Engineering, University of Bergamo, 24044 Dalmine, BG, Italy
| | - Karine Elihn
- Department of Environmental Science, Stockholm University, 11419 Stockholm, Sweden
| | - Benjamin Piña
- Institute of Environmental Assessment and Water Research, CSIC, 08034 Barcelona, Spain.
| |
Collapse
|
2
|
Su D, Luo X, Chen J, Lu N, Zhao J, Wan Y, Gao Y, Liu Q, Luo Z. Construction of a three-dimensional inflammation model of human bronchial epithelial cells BEAS-2B by using the self-assembling D-form peptide Sciobio-Ⅲ. Biochem Biophys Res Commun 2024; 704:149701. [PMID: 38408415 DOI: 10.1016/j.bbrc.2024.149701] [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: 11/13/2023] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 02/28/2024]
Abstract
Human bronchial epithelial cells in the airway system, as the primary barrier between humans and the surrounding environment, assume a crucial function in orchestrating the processes of airway inflammation. Target to develop a new three-dimensional (3D) inflammatory model to airway system, and here we report a strategy by using self-assembling D-form peptide to cover the process. By testing physicochemical properties and biocompatibility of Sciobio-Ⅲ, we confirmed that it can rapidly self-assembles under the trigger of ions to form a 3D nanonetwork-like scaffold, which supports 3D cell culture including the cell strains like BEAS-2B cells. Subsequently, inflammation model was established by lipopolysaccharide (LPS), the expression of some markers of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and interleukin-8 (IL-8), the levels of relevant inflammatory factors were measured by RT-qPCR and the secretion profile of inflammatory cytokines by ELISA, are obtained the quite difference effects in 2D and 3D microenvironment, which suggested Sciobio-Ⅲ hydrogel is an ideal scaffold that create the microenvironment for 3D cell culture. Here we are success to establish a 3D inflammation model for airway system. This innovative model allows for rapid and accurate evaluation of drug metabolism and toxicological side effects, hope to use in drug screening for airway inflammatory diseases and beyond.
Collapse
Affiliation(s)
- Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China; Department of Materials Science and Engineering, University of California, Irvine, CA, 92697, USA
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Jiawei Zhao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China; Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, IA, 52242, USA
| | - Yu Gao
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Qichen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Dichtl S, Posch W, Wilflingseder D. The breathtaking world of human respiratory in vitro models: Investigating lung diseases and infections in 3D models, organoids, and lung-on-chip. Eur J Immunol 2024; 54:e2250356. [PMID: 38361030 DOI: 10.1002/eji.202250356] [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: 03/31/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 02/17/2024]
Abstract
The COVID-19 pandemic illustrated an urgent need for sophisticated, human tissue models to rapidly test and develop effective treatment options against this newly emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Thus, in particular, the last 3 years faced an extensive boost in respiratory and pulmonary model development. Nowadays, 3D models, organoids and lung-on-chip, respiratory models in perfusion, or precision-cut lung slices are used to study complex research questions in human primary cells. These models provide physiologically relevant systems for studying SARS-CoV-2 and, of course, other respiratory pathogens, but they are, too, suited for studying lung pathologies, such as CF, chronic obstructive pulmonary disease, or asthma, in more detail in terms of viral infection. With these models, the cornerstone has been laid for further advancing the organs by, for example, inclusion of several immune cell types or humoral immune components, combination with other organs in microfluidic organ-on-chip devices, standardization and harmonization of the devices for reliable and reproducible drug and vaccine testing in high throughput.
Collapse
Affiliation(s)
- Stefanie Dichtl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wilfried Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
4
|
Lutsch CT, Feng L, Gómez Hohn A, Brandt L, Tamm S, Janciauskiene S, Stanke F, Jonigk D, Dittrich AM, Braubach P. A Fast Scoring of Human Primary Respiratory Epithelia Grown at Air-Liquid Interface (ALI) to Assess Epithelial Morphology in Research and Personalized Medicine Settings. J Pers Med 2024; 14:109. [PMID: 38248810 PMCID: PMC10817428 DOI: 10.3390/jpm14010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/29/2023] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND In recent years, increasingly complex ALI protocols involving specialized, albeit laboratory-specific media have been established, while at the same time, many studies compile the data of only a few ALI donors in spite of site-, protocol- and donor-specific differentiation. METHODS We describe a simple morphology scoring protocol using histology material derived from epithelia grown on ALI inserts in parallel to other, more complex readouts. RESULTS Among more than 100 ALI inserts derived from different donors, significant differences in layer score (p = 0.001) and goblet cell score (p = 0.002) were observed when ALI epithelia derived from explanted lung material were compared to trachea-derived ALI cultures. Cortisol withdrawal for the final 2 days of ALI cultures influenced goblet cell density (p = 0.001). CONCLUSIONS While the histology score provides less resolution than FACS- or OMICs- based single cell analyses, the use of a subportion of the ALI epithelia grown on inserts makes it feasible to combine morphology assessment and other readouts of the same insert. This allows us to control for basic ALI morphology in research and personalized medicine settings in order to assess and, if desired, control for the impact of ALI culture protocols, site- and donor-specific influences on outcome of studies of ALI-derived epithelia.
Collapse
Affiliation(s)
- Christopher T. Lutsch
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
| | - Longhua Feng
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
| | - Ana Gómez Hohn
- Institute for Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Lennart Brandt
- Institute for Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Stephanie Tamm
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
| | - Sabina Janciauskiene
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Frauke Stanke
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
- Institute of Pathology, School of Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany; (S.J.); (D.J.)
| | - Peter Braubach
- Institute for Pathology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
5
|
Sharma M, Stucki AO, Verstraelen S, Stedeford TJ, Jacobs A, Maes F, Poelmans D, Van Laer J, Remy S, Frijns E, Allen DG, Clippinger AJ. Human cell-based in vitro systems to assess respiratory toxicity: a case study using silanes. Toxicol Sci 2023; 195:213-230. [PMID: 37498623 PMCID: PMC10535780 DOI: 10.1093/toxsci/kfad074] [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] [Indexed: 07/28/2023] Open
Abstract
Inhalation is a major route by which human exposure to substances can occur. Resources have therefore been dedicated to optimize human-relevant in vitro approaches that can accurately and efficiently predict the toxicity of inhaled chemicals for robust risk assessment and management. In this study-the IN vitro Systems to PredIct REspiratory toxicity Initiative-2 cell-based systems were used to predict the ability of chemicals to cause portal-of-entry effects on the human respiratory tract. A human bronchial epithelial cell line (BEAS-2B) and a reconstructed human tissue model (MucilAir, Epithelix) were exposed to triethoxysilane (TES) and trimethoxysilane (TMS) as vapor (mixed with N2 gas) at the air-liquid interface. Cell viability, cytotoxicity, and secretion of inflammatory markers were assessed in both cell systems and, for MucilAir tissues, morphology, barrier integrity, cilia beating frequency, and recovery after 7 days were also examined. The results show that both cell systems provide valuable information; the BEAS-2B cells were more sensitive in terms of cell viability and inflammatory markers, whereas MucilAir tissues allowed for the assessment of additional cellular effects and time points. As a proof of concept, the data were also used to calculate human equivalent concentrations. As expected, based on chemical properties and existing data, the silanes demonstrated toxicity in both systems with TMS being generally more toxic than TES. Overall, the results demonstrate that these in vitro test systems can provide valuable information relevant to predicting the likelihood of toxicity following inhalation exposure to chemicals in humans.
Collapse
Affiliation(s)
- Monita Sharma
- PETA Science Consortium International e.V., 70499 Stuttgart, Germany
| | - Andreas O Stucki
- PETA Science Consortium International e.V., 70499 Stuttgart, Germany
| | - Sandra Verstraelen
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | | | - An Jacobs
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - Frederick Maes
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - David Poelmans
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - Jo Van Laer
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - Sylvie Remy
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - Evelien Frijns
- Sustainable HEALTH Unit, Flemish Institute for Technological Research (VITO), BE-2400 Mol, Belgium
| | - David G Allen
- Inotiv, Research Triangle Park, North Carolina 27560, USA
| | - Amy J Clippinger
- PETA Science Consortium International e.V., 70499 Stuttgart, Germany
| |
Collapse
|
6
|
Wang K, Yu Y, Han R, Wang X, Zhao Y, Tang H, Li G. [Establishment of a culture system for human nasal mucosa organoids with controllable differentiation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:868-877. [PMID: 35790437 DOI: 10.12122/j.issn.1673-4254.2022.06.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To establish a culture system for human nasal mucosal organoids with controllable differentiation to reproduce the structure and function of the source tissue through staged expansion-differentiation culture. METHODS Fresh samples of surgically resected middle turbinate and nasal polyp tissues were collected, from which the nasal mucosa epithelial cells were isolated by enzymatic digestion and filtration for continuous culture at the air-liquid interface for expansion (EO group) or staged culture for expansion and differentiation (DO group). Immunohistochemical staining was used to characterize the structure, cellular composition and ciliary function of nasal mucosal organoids in the two groups. The secretion function of the differentiated nasal mucosal organoids in DO group was evaluated using PAS staining. RESULTS Both of the two organoid culture systems yielded vacuolar or solid spherical 3D organoids, and their diameters increased progressively with time. On day 16 of culture, more vacuolar organoids occurred in DO group, while more solid spherical organoids were seen in EO group, and the proportion of vacuoles was significantly greater in DO group than in EO group [(54.67±13.26)% vs (21.67±8.57)%, P < 0.05]. Short tandem repeat (STR) test of the nasal mucosal organoids and the source tissue showed a 100% match between them. On day 21 of culture, scanning and transmission electron microscopy of the nasal mucosal organoids identified ultrastructure of cilia in DO group and short villi structure in most of the organoids in EO group. Immunohistochemical staining showed positivity for P63 (basal cells), β-tubulin (ciliated columnar cells), and MUC5AC (goblet cells) in the organoids. Compared with those in EO group, the organoids in DO group showed significantly greater percentages of ciliated cells [(7.95±1.81)% vs (27.04±5.91)%, P < 0.05] and goblet cells [(14.46±0.93)% vs (39.85±5.43)%, P < 0.05) with a similar percentage of basal cells [(56.91±14.12)% vs (53.42±15.77)%, P > 0.05]. The differentiated nasal mucosal organoids in DO group were positively stained for glycogen. CONCLUSION The staged expansion-differentiation culture method allows more stable and prolonged growth of the cultured cells in vitro to produce organoids with controllable differentiation closely resembling the morphological structure and functions (ciliary function and secretory function) of the source tissue.
Collapse
Affiliation(s)
- K Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Yu
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - R Han
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - X Wang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Y Zhao
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - H Tang
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - G Li
- Department of Otolaryngology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Czechtizky W, Su W, Ripa L, Schiesser S, Höijer A, Cox RJ. Advances in the design of new types of inhaled medicines. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:93-162. [PMID: 35753716 DOI: 10.1016/bs.pmch.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Inhalation of small molecule drugs has proven very efficacious for the treatment of respiratory diseases due to enhanced efficacy and a favourable therapeutic index compared with other dosing routes. It enables targeted delivery to the lung with rapid onset of therapeutic action, low systemic drug exposure, and thereby reduced systemic side effects. An increasing number of pharmaceutical companies and biotechs are investing in new modalities-for this review defined as therapeutic molecules with a molecular weight >800Da and therefore beyond usual inhaled small molecule drug-like space. However, our experience with inhaled administration of PROTACs, peptides, oligonucleotides (antisense oligonucleotides, siRNAs, miRs and antagomirs), diverse protein scaffolds, antibodies and antibody fragments is still limited. Investigating the retention and metabolism of these types of molecules in lung tissue and fluid will contribute to understanding which are best suited for inhalation. Nonetheless, the first such therapeutic molecules have already reached the clinic. This review will provide information on the physiology of healthy and diseased lungs and their capacity for drug metabolism. It will outline the stability, aggregation and immunogenicity aspects of new modalities, as well as recap on formulation and delivery aspects. It concludes by summarising clinical trial outcomes with inhaled new modalities based on information available at the end of 2021.
Collapse
Affiliation(s)
- Werngard Czechtizky
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden.
| | - Wu Su
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Lena Ripa
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Stefan Schiesser
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Andreas Höijer
- Cardiovascular, Renal & Metabolism CMC Projects, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Rhona J Cox
- Department of Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal & Metabolism, BioPharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
8
|
Sakolish C, Georgescu A, Huh DD, Rusyn I. A model of human small airway on a chip for studies of sub-acute effects of inhalation toxicants. Toxicol Sci 2022; 187:267-278. [PMID: 35357501 DOI: 10.1093/toxsci/kfac036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Testing for acute inhalation hazards is conducted in animals; however, a number of robust in vitro human cell-based alternatives were developed and tested. These models range in complexity from cultures of cell lines or primary cells in air-liquid interface on trans-wells, to more complex and physiologically-relevant flow- and mechanical stimulation-enabled tissue chips. While the former models are relatively straightforward to establish and can be tested in medium/high-throughput, the latter require specialized equipment and lack in throughput. In this study, we developed a device that can be easily manufactured while allowing for the production of a differentiated lung tissue. This multilayered microfluidic device enables co-culture of primary human small airway epithelial cells and lung microvascular endothelial cells under physiological conditions for up to 14 days and recreates the parenchymal-vascular interface in the distal lung. To explore the potential of this airway-on-a-chip for applications in inhalation toxicology, we also devised a system that allows for direct gas/aerosol exposures of the engineered airway epithelium to noxious stimuli known to cause adverse respiratory effects, including dry flowing air, lipopolysaccharide, particulate matter, and iodomethane. This study generated quantitative, high-content data that were indicative of aberrant changes in biochemical (lactate dehydrogenase), barrier (dextran permeability), functional (ciliary beating), and molecular (imaging for various markers) phenotypes of the small airway epithelium due to inhalational exposures. This study is significant because it established an in vitro model of human small airway on a chip that can be used in medium/high-throughput studies of sub-acute effects of inhalation toxicants.
Collapse
Affiliation(s)
- Courtney Sakolish
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843, USA
| | - Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
9
|
Meldrum K, Evans SJ, Vogel U, Tran L, Doak SH, Clift MJD. The influence of exposure approaches to in vitro lung epithelial barrier models to assess engineered nanomaterial hazard. Nanotoxicology 2022; 16:114-134. [PMID: 35343373 DOI: 10.1080/17435390.2022.2051627] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exposure to engineered nanomaterials (ENM) poses a potential health risk to humans through long-term, repetitive low-dose exposures. Currently, this is not commonplace within in vitro lung cell cultures. Therefore, the purpose of this study was to consider the optimal exposure approach toward determining the stability, sensitivity and validity of using in vitro lung cell mono- and co-cultures to determine ENM hazard. A range of exposure scenarios were conducted with DQ12 (previously established as a positive particle control) (historic and re-activated), TiO2 (JRC NM-105) and BaSO4 (JRC NM-220) on both monocultures of A549 cells as well as co-cultures of A549 cells and differentiated THP-1 cells. Cell cultures were exposed to either a single, or a repeated exposure over 24, 48- or 72-hours at in vivo extrapolated concentrations of 0-5.2 µg/cm2, 0-6 µg/cm2 and 0-1µg/cm2. The focus of this study was the pro-inflammatory, cytotoxic and genotoxic response elicited by these ENMs. Exposure to DQ12 caused pro-inflammatory responses after 48 hours repeat exposures, as well as increases in micronucleus frequency. Neither TiO2 nor BaSO4 elicited a pro-inflammatory response at this time point. However, there was induction of IL-6 after 24 hours TiO2 exposure. In conclusion, it is important to consider the appropriateness of the positive control implemented, the cell culture model, the time of exposure as well as the type of exposure (bolus or fractionated) before establishing if an in vitro model is appropriate to determine the level of response to the specific ENM of interest.
Collapse
Affiliation(s)
- Kirsty Meldrum
- In Vitro Toxicology Group, Swansea University, Swansea, UK
| | | | - Ulla Vogel
- The National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Lang Tran
- Institute of Occupational Medicine (IOM), Edinburgh, UK
| | - Shareen H Doak
- In Vitro Toxicology Group, Swansea University, Swansea, UK
| | | |
Collapse
|
10
|
Juarez-Facio AT, Castilla C, Corbière C, Lavanant H, Afonso C, Morin C, Merlet-Machour N, Chevalier L, Vaugeois JM, Yon J, Monteil C. Development of a standardized in vitro approach to evaluate microphysical, chemical, and toxicological properties of combustion-derived fine and ultrafine particles. J Environ Sci (China) 2022; 113:104-117. [PMID: 34963520 DOI: 10.1016/j.jes.2021.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 06/14/2023]
Abstract
Ultrafine particles represent a growing concern in the public health community but their precise role in many illnesses is still unknown. This lack of knowledge is related to the experimental difficulty in linking their biological effects to their multiple properties, which are important determinants of toxicity. Our aim is to propose an interdisciplinary approach to study fine (FP) and ultrafine (UFP) particles, generated in a controlled manner using a miniCAST (Combustion Aerosol Standard) soot generator used with two different operating conditions (CAST1 and CAST3). The chemical characterization was performed by an untargeted analysis using ultra-high resolution mass spectrometry. In conjunction with this approach, subsequent analysis by gas chromatography-mass spectrometry (GC-MS) was performed to identify polycyclic aromatic hydrocarbons (PAH). CAST1 enabled the generation of FP with a predominance of small PAH molecules, and CAST3 enabled the generation of UFP, which presented higher numbers of carbon atoms corresponding to larger PAH molecules. Healthy normal human bronchial epithelial (NHBE) cells differentiated at the air-liquid interface (ALI) were directly exposed to these freshly emitted FP and UFP. Expression of MUC5AC, FOXJ1, OCLN and ZOI as well as microscopic observation confirmed the ciliated pseudostratified epithelial phenotype. Study of the mass deposition efficiency revealed a difference between the two operating conditions, probably due to the morphological differences between the two categories of particles. We demonstrated that only NHBE cells exposed to CAST3 particles induced upregulation in the gene expression of IL-8 and NQO1. This approach offers new perspectives to study FP and UFP with stable and controlled properties.
Collapse
Affiliation(s)
| | - Clément Castilla
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | | | - Hélène Lavanant
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | - Carlos Afonso
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | - Christophe Morin
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, COBRA, 76000 Rouen, France
| | | | - Laurence Chevalier
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, GPM-UMR6634, 76000 Rouen, France
| | | | - Jérôme Yon
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, CORIA, 76000 Rouen, France
| | | |
Collapse
|
11
|
Wu W, Choi EJ, Wang B, Zhang K, Adam A, Huang G, Tunkle L, Huang P, Goru R, Imirowicz I, Henry L, Lee I, Dong J, Wang T, Bao X. Changes of Small Non-coding RNAs by Severe Acute Respiratory Syndrome Coronavirus 2 Infection. Front Mol Biosci 2022; 9:821137. [PMID: 35281271 PMCID: PMC8905365 DOI: 10.3389/fmolb.2022.821137] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/19/2022] [Indexed: 01/11/2023] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19), which results from the rapid spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a significant global public health threat, with molecular mechanisms underlying its pathogenesis largely unknown. In the context of viral infections, small non-coding RNAs (sncRNAs) are known to play important roles in regulating the host responses, viral replication, and host-virus interaction. Compared with other subfamilies of sncRNAs, including microRNAs (miRNAs) and Piwi-interacting RNAs (piRNAs), tRNA-derived RNA fragments (tRFs) are relatively new and emerge as a significant regulator of host-virus interactions. Using T4 PNK-RNA-seq, a modified next-generation sequencing (NGS), we found that sncRNA profiles in human nasopharyngeal swabs (NPS) samples are significantly impacted by SARS-CoV-2. Among impacted sncRNAs, tRFs are the most significantly affected and most of them are derived from the 5'-end of tRNAs (tRF5). Such a change was also observed in SARS-CoV-2-infected airway epithelial cells. In addition to host-derived ncRNAs, we also identified several small virus-derived ncRNAs (svRNAs), among which a svRNA derived from CoV2 genomic site 346 to 382 (sv-CoV2-346) has the highest expression. The induction of both tRFs and sv-CoV2-346 has not been reported previously, as the lack of the 3'-OH ends of these sncRNAs prevents them to be detected by routine NGS. In summary, our studies demonstrated the involvement of tRFs in COVID-19 and revealed new CoV2 svRNAs.
Collapse
Affiliation(s)
- Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, United States
| | - Eun-Jin Choi
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, United States
| | - Binbin Wang
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Ke Zhang
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, United States
| | - Awadalkareem Adam
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Gengming Huang
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Leo Tunkle
- miRcore, Ann Arbor, MI, United States
- Department of Nuclear Engineering and Radiological Sience, University of Michigan, Ann Arbor, MI, United States
- Department of Computer Science, University of Michigan, Ann Arbor, MI, United States
| | - Philip Huang
- miRcore, Ann Arbor, MI, United States
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Rohit Goru
- miRcore, Ann Arbor, MI, United States
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Isabella Imirowicz
- miRcore, Ann Arbor, MI, United States
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Leanne Henry
- miRcore, Ann Arbor, MI, United States
- Department of Computer Science, University of Michigan, Ann Arbor, MI, United States
| | - Inhan Lee
- miRcore, Ann Arbor, MI, United States
| | - Jianli Dong
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, United States
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX, United States
| | - Tian Wang
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, United States
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX, United States
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, United States
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX, United States
- The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
12
|
In Vitro–In Silico Modeling of Caffeine and Diclofenac Permeation in Static and Fluidic Systems with a 16HBE Lung Cell Barrier. Pharmaceuticals (Basel) 2022; 15:ph15020250. [PMID: 35215362 PMCID: PMC8876625 DOI: 10.3390/ph15020250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Static in vitro permeation experiments are commonly used to gain insights into the permeation properties of drug substances but exhibit limitations due to missing physiologic cell stimuli. Thus, fluidic systems integrating stimuli, such as physicochemical fluxes, have been developed. However, as fluidic in vitro studies display higher complexity compared to static systems, analysis of experimental readouts is challenging. Here, the integration of in silico tools holds the potential to evaluate fluidic experiments and to investigate specific simulation scenarios. This study aimed to develop in silico models that describe and predict the permeation and disposition of two model substances in a static and fluidic in vitro system. For this, in vitro permeation studies with a 16HBE cellular barrier under both static and fluidic conditions were performed over 72 h. In silico models were implemented and employed to describe and predict concentration–time profiles of caffeine and diclofenac in various experimental setups. For both substances, in silico modeling identified reduced apparent permeabilities in the fluidic compared to the static cellular setting. The developed in vitro–in silico modeling framework can be expanded further, integrating additional cell tissues in the fluidic system, and can be employed in future studies to model pharmacokinetic and pharmacodynamic drug behavior.
Collapse
|
13
|
Jung O, Tung YT, Sim E, Chen YC, Lee E, Ferrer M, Song MJ. Development of human-derived, three-dimensional respiratory epithelial tissue constructs with perfusable microvasculature on a high-throughput microfluidics screening platform. Biofabrication 2022; 14. [PMID: 35166694 PMCID: PMC10053540 DOI: 10.1088/1758-5090/ac32a5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/22/2021] [Indexed: 11/12/2022]
Abstract
The COVID-19 pandemic has highlighted the need for human respiratory tract-based assay platforms for efficient discovery and development of antivirals and disease-modulating therapeutics. Physiologically relevant tissue models of the lower respiratory tract (LRT), including the respiratory bronchioles and the alveolar sacs, are of high interest because they are the primary site of severe SARS-CoV-2 infection and are most affected during the terminal stage of COVID-19. Current epithelial lung models used to study respiratory viral infections include lung epithelial cells at the air-liquid interface (ALI) with fibroblasts and endothelial cells, but such models do not have a perfusable microvascular network to investigate both viral infectivity and viral infection-induced thrombotic events. Using a high throughput, 64-chip microfluidic plate-based platform, we have developed two novel vascularized, LRT multi-chip models for the alveoli and the small airway. Both models include a perfusable microvascular network consisting of human primary microvascular endothelial cells, fibroblasts and pericytes. The established biofabrication protocols also enable the formation of differentiated lung epithelial layers at the ALI on top of the vascularized tissue bed. We validated the physiologically relevant cellular composition, architecture and perfusion of the vascularized lung tissue models using fluorescence microscopy, flow cytometry, and electrical resistance measurements. These vascularized, perfusable microfluidic lung tissue on high throughput assay platforms will enable the development of respiratory viral infection and disease models for research investigation and drug discovery.
Collapse
Affiliation(s)
- Olive Jung
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America.,Biomedical Ultrasonics, Biotherapy and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Yen-Ting Tung
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Esther Sim
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Yu-Chi Chen
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Emily Lee
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| | - Min Jae Song
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, United States of America
| |
Collapse
|
14
|
Campos CL, Gomes LR, Covarrubias AE, Kato EE, Souza GG, Vasconcellos SA, Heinemann MB, Martins EAL, Ho PL, Da Costa RMA, Da Silva JB. A Three-Dimensional Lung Cell Model to Leptospira Virulence Investigations. Curr Microbiol 2022; 79:57. [PMID: 34982247 DOI: 10.1007/s00284-021-02720-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/08/2021] [Indexed: 11/30/2022]
Abstract
Leptospirosis is a worldwide zoonosis and a serious public health threat in tropical and subtropical areas. The etiologic agents of leptospirosis are pathogenic spirochetes from the genus Leptospira. In severe cases, patients develop a pulmonary hemorrhage that is associated with high fatality rates. Several animal models were established for leptospirosis studies, such as rodents, dogs, and monkeys. Although useful to study the relationship among Leptospira and its hosts, the animal models still exhibit economic and ethical limitation reasons and do not fully represent the human infection. As an attempt to bridge the gap between animal studies and clinical information from patients, we established a three-dimensional (3-D) human lung cell culture for Leptospira infection. We show that Leptospira is able to efficiently infect the cell lung spheroids and also to infiltrate in deeper areas of the cell aggregates. The ability to infect the 3-D lung cell aggregates was time-dependent. The 3-D spheroids infection occurred up to 120 h in studies with two serovars, Canicola and Copenhageni. We standardized the number of bacteria in the initial inoculum for infection of the spheroids and we also propose two alternative culture media conditions. This new approach was validated by assessing the expression of three genes of Leptospira related to virulence and motility. The transcripts of these genes increased in both culture conditions, however, in higher rates and earlier times in the 3-D culture. We also assessed the production of chemokines by the 3-D spheroids before and after Leptospira infection, confirming induction of two of them, mainly in the 3-D spheroids. Chemokine CCL2 was expressed only in the 3-D cell culture. Increasing of this chemokine was observed previously in infected animal models. This new approach provides an opportunity to study the interaction of Leptospira with the human lung epithelium in vitro.
Collapse
Affiliation(s)
- Camila L Campos
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
| | - Luciana R Gomes
- Laboratório de Ciclo Celular-Center for Research on Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Ambart E Covarrubias
- Facultad de Ciencias de la Salud, Escuela de Tecnología Médica, Universidad San Sebastian, Concepción, Chile
| | - Ellen E Kato
- Laboratorio de Fisiopatologia, Instituto Butantan, São Paulo, Brazil
| | - Gisele G Souza
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Silvio A Vasconcellos
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Marcos B Heinemann
- Laboratório de Zoonoses Bacterianas, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | | | - Paulo L Ho
- Divisão BioIndustrial, Instituto Butantan, São Paulo, Brazil
| | - Renata M A Da Costa
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.,Global Antibiotics Research and Development Partnership (GARDP), Chemin Louis-Dunant 15, 1202, Geneva, Switzerland
| | - Josefa B Da Silva
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil.
| |
Collapse
|
15
|
Moreira A, Müller M, Costa PF, Kohl Y. Advanced In Vitro Lung Models for Drug and Toxicity Screening: The Promising Role of Induced Pluripotent Stem Cells. Adv Biol (Weinh) 2021; 6:e2101139. [PMID: 34962104 DOI: 10.1002/adbi.202101139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/25/2021] [Indexed: 12/24/2022]
Abstract
The substantial socioeconomic burden of lung diseases, recently highlighted by the disastrous impact of the coronavirus disease 2019 (COVID-19) pandemic, accentuates the need for interventive treatments capable of decelerating disease progression, limiting organ damage, and contributing to a functional tissue recovery. However, this is hampered by the lack of accurate human lung research models, which currently fail to reproduce the human pulmonary architecture and biochemical environment. Induced pluripotent stem cells (iPSCs) and organ-on-chip (OOC) technologies possess suitable characteristics for the generation of physiologically relevant in vitro lung models, allowing for developmental studies, disease modeling, and toxicological screening. Importantly, these platforms represent potential alternatives for animal testing, according to the 3Rs (replace, reduce, refine) principle, and hold promise for the identification and approval of new chemicals under the European REACH (registration, evaluation, authorization and restriction of chemicals) framework. As such, this review aims to summarize recent progress made in human iPSC- and OOC-based in vitro lung models. A general overview of the present applications of in vitro lung models is presented, followed by a summary of currently used protocols to generate different lung cell types from iPSCs. Lastly, recently developed iPSC-based lung models are discussed.
Collapse
Affiliation(s)
| | - Michelle Müller
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany
| | - Pedro F Costa
- BIOFABICS, Rua Alfredo Allen 455, Porto, 4200-135, Portugal
| | - Yvonne Kohl
- Department of Bioprocessing and Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280, Sulzbach, Germany.,Postgraduate Course for Toxicology and Environmental Toxicology, Medical Faculty, University of Leipzig, Johannisallee 28, 04103, Leipzig, Germany
| |
Collapse
|
16
|
Wu W, Choi EJ, Wang B, Zhang K, Adam A, Huang G, Tunkle L, Huang P, Goru R, Imirowicz I, Henry L, Lee I, Dong J, Wang T, Bao X. Changes of small non-coding RNAs by severe acute respiratory syndrome coronavirus 2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34981063 DOI: 10.1101/2021.12.16.472982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19), which results from the rapid spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a significant global public health threat, with molecular mechanisms underlying its pathogenesis largely unknown. Small non-coding RNAs (sncRNAs) are known to play important roles in almost all biological processes. In the context of viral infections, sncRNAs have been shown to regulate the host responses, viral replication, and host-virus interaction. Compared with other subfamilies of sncRNAs, including microRNAs (miRNAs) and Piwi-interacting RNAs (piRNAs), tRNA-derived RNA fragments (tRFs) are relatively new and emerge as a significant regulator of host-virus interactions. Using T4 PNK-RNA-seq, a modified next-generation sequencing (NGS), we recently found that nasopharyngeal swabs (NPS) samples from SARS-CoV-2 positive and negative subjects show a significant difference in sncRNA profiles. There are about 166 SARS-CoV-2-impacted sncRNAs. Among them, tRFs are the most significantly affected and almost all impacted tRFs are derived from the 5'-end of tRNAs (tRF5). Using a modified qRT-PCR, which was recently developed to specifically quantify tRF5s by isolating the tRF signals from its corresponding parent tRNA signals, we validated that tRF5s derived from tRNA GluCTC (tRF5-GluCTC), LysCTT (tRF5-LysCTT), ValCAC (tRF5-ValCAC), CysGCA (tRF5-CysGCA) and GlnCTG (tRF5-GlnCTG) are enhanced in NPS samples of SARS-CoV2 patients and SARS-CoV2-infected airway epithelial cells. In addition to host-derived ncRNAs, we also identified several sncRNAs derived from the virus (svRNAs), among which a svRNA derived from CoV2 genomic site 346 to 382 (sv-CoV2-346) has the highest expression. The induction of both tRFs and sv-CoV2-346 has not been reported previously, as the lack of the 3'-OH ends of these sncRNAs prevents them to be detected by routine NGS. In summary, our studies demonstrated the involvement of tRFs in COVID-19 and revealed new CoV2 svRNAs.
Collapse
|
17
|
Juarez Facio AT, Yon J, Corbière C, Rogez-Florent T, Castilla C, Lavanant H, Mignot M, Devouge-Boyer C, Logie C, Chevalier L, Vaugeois JM, Monteil C. Toxicological impact of organic ultrafine particles (UFPs) in human bronchial epithelial BEAS-2B cells at air-liquid interface. Toxicol In Vitro 2021; 78:105258. [PMID: 34653646 DOI: 10.1016/j.tiv.2021.105258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/30/2021] [Accepted: 10/09/2021] [Indexed: 12/26/2022]
Abstract
Air pollution has significant health effects worldwide, and airborne particles play a significant role in these effects. Ultrafine particles (UFPs) have an aerodynamic diameter of 0.1 μm or less, can penetrate deep into the respiratory tree, and are more toxic due to their large specific surface area, which should adsorb organic compounds. The aim of this study is to show the toxicological effects of UFPs with high organic content at low dose on BEAS-2B cells through at air-liquid interface (ALI) exposure using a Vitrocell® technology and a miniCAST (Combustion Aerosol Standard) generator. In conjunction with this approach, chemical analysis of particles and gas phase was performed to evaluate the presence of polycyclic aromatic hydrocarbons (PAHs). Chemical analyses confirmed the presence of PAHs in UFPs. With this experimental setup, exposure of the BEAS-2B cells induced neither cytotoxicity nor mitochondrial dysfunction. However, an increase of oxidative stress was observed, as assessed through Nrf2, NQO1, HO-1, CuZnSOD, MnSOD, and Catalase gene expression, together with significant induction of genes related to xenobiotic metabolism CYP1A1 and CYP1B1. Negative regulation of inflammatory genes expression (IL-6 and IL-8) was present three hours after the exposition to the UFPs. Taken together, this experimental approach, using repeatable conditions, should help to clarify the mechanisms by which organic UFPs induce toxicological effects.
Collapse
Affiliation(s)
| | - J Yon
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, CORIA, 76000 Rouen, France
| | - C Corbière
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 76000 Rouen, France
| | | | - C Castilla
- Normandie Univ, INSA Rouen, UMR 6014 CNRS, COBRA, 76801, Saint Etienne Du Rouvray, France
| | - H Lavanant
- Normandie Univ, INSA Rouen, UMR 6014 CNRS, COBRA, 76801, Saint Etienne Du Rouvray, France
| | - M Mignot
- Normandie Univ, INSA Rouen, UMR 6014 CNRS, COBRA, 76801, Saint Etienne Du Rouvray, France
| | - C Devouge-Boyer
- Normandie Univ, INSA Rouen, UMR 6014 CNRS, COBRA, 76801, Saint Etienne Du Rouvray, France
| | - C Logie
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 76000 Rouen, France
| | - L Chevalier
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, GPM-UMR6634, 76000 Rouen, France
| | - J-M Vaugeois
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 76000 Rouen, France
| | - C Monteil
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 76000 Rouen, France.
| |
Collapse
|
18
|
Duan W, Cen Y, Lin C, Ouyang H, Du K, Kumar A, Wang B, Avolio J, Grasemann H, Moraes TJ. Inflammatory epithelial cytokines after in vitro respiratory syncytial viral infection are associated with reduced lung function. ERJ Open Res 2021; 7:00365-2021. [PMID: 34527729 PMCID: PMC8435810 DOI: 10.1183/23120541.00365-2021] [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: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 11/27/2022] Open
Abstract
Respiratory syncytial virus (RSV) infections in early life predispose children with cystic fibrosis (CF) to more severe lung function decline in later life. The mechanisms explaining the associations between RSV and progression of CF lung disease are not clear. In this study, a human bronchial epithelial cell line and primary human nasal epithelial cells (PNECs) from individuals with CF and healthy control donors were infected with RSV. Real-time PCR, plaque assay, cytokine detection, immunofluorescence and Western blot analyses were performed. RSV is replicated to a higher degree in CF epithelial cells as compared to control cells; however, no defects in innate immune pathways were identified in CF cells. Rather, primary p.Phe508del cystic fibrosis transmembrane conductance regulator PNECs produced more cytokines after RSV infection than control cells. Moreover, interleukin-8 and tumour necrosis factor-α production post RSV negatively correlated with lung function (% predicted forced expiratory volume in 1 s) in the individuals who donated the cells. These data suggest that CF epithelium has a dysfunctional response to RSV allowing for enhanced viral replication and an exaggerated inflammatory response that ultimately may predispose to greater airway inflammation and reduced lung function. This work demonstrates an association between epithelial inflammatory cytokines after in vitro viral infection and lung function in cystic fibrosis, and reinforces the importance of studying innate immune epithelial cell function in cystic fibrosishttps://bit.ly/3gDNwwo
Collapse
Affiliation(s)
- Wenming Duan
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Yuchen Cen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Dept of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Cindy Lin
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Hong Ouyang
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Kai Du
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Anushree Kumar
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Borui Wang
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Julie Avolio
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Hartmut Grasemann
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Division of Respiratory Medicine, Dept of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.,Dept of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Respiratory Medicine, Dept of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
19
|
Artzy-Schnirman A, Arber Raviv S, Doppelt Flikshtain O, Shklover J, Korin N, Gross A, Mizrahi B, Schroeder A, Sznitman J. Advanced human-relevant in vitro pulmonary platforms for respiratory therapeutics. Adv Drug Deliv Rev 2021; 176:113901. [PMID: 34331989 PMCID: PMC7611797 DOI: 10.1016/j.addr.2021.113901] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 02/08/2023]
Abstract
Over the past years, advanced in vitro pulmonary platforms have witnessed exciting developments that are pushing beyond traditional preclinical cell culture methods. Here, we discuss ongoing efforts in bridging the gap between in vivo and in vitro interfaces and identify some of the bioengineering challenges that lie ahead in delivering new generations of human-relevant in vitro pulmonary platforms. Notably, in vitro strategies using foremost lung-on-chips and biocompatible "soft" membranes have focused on platforms that emphasize phenotypical endpoints recapitulating key physiological and cellular functions. We review some of the most recent in vitro studies underlining seminal therapeutic screens and translational applications and open our discussion to promising avenues of pulmonary therapeutic exploration focusing on liposomes. Undeniably, there still remains a recognized trade-off between the physiological and biological complexity of these in vitro lung models and their ability to deliver assays with throughput capabilities. The upcoming years are thus anticipated to see further developments in broadening the applicability of such in vitro systems and accelerating therapeutic exploration for drug discovery and translational medicine in treating respiratory disorders.
Collapse
Affiliation(s)
- Arbel Artzy-Schnirman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Sivan Arber Raviv
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | | | - Jeny Shklover
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Adi Gross
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Boaz Mizrahi
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Avi Schroeder
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel.
| |
Collapse
|
20
|
Arezki Y, Cornacchia J, Rapp M, Lebeau L, Pons F, Ronzani C. A Co-Culture Model of the Human Respiratory Tract to Discriminate the Toxicological Profile of Cationic Nanoparticles According to Their Surface Charge Density. TOXICS 2021; 9:210. [PMID: 34564361 PMCID: PMC8470030 DOI: 10.3390/toxics9090210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 12/16/2022]
Abstract
This study aimed at discriminating with sensitivity the toxicological effects of carbon dots (CDs) with various zeta potential (ζ) and charge density (Qek) in different cellular models of the human respiratory tract. One anionic and three cationic CDs were synthetized as follows: CD-COOH (ζ = -43.3 mV); CD-PEI600 (Qek = 4.70 µmol/mg; ζ = +31.8 mV); CD-PEHA (Qek = 3.30 µmol/mg; ζ = +29.2 mV) and CD-DMEDA (Qek = 0.01 µmol/mg; ζ = +11.1 mV). Epithelial cells (A549) and macrophages (THP-1) were seeded alone or as co-cultures with different A549:THP-1 ratios. The obtained models were characterized, and multiple biological responses evoked by CDs were assessed in the mono-cultures and the best co-culture model. With 14% macrophages, the 2:1 ratio co-culture best mimicked the in vivo conditions and responded to lipopolysaccharides. The anionic CD did not induce any effect in the mono-cultures nor in the co-culture. Among the cationic CDs, the one with the highest charge density (CD-PEI600) induced the most pronounced responses whatever the culture model. The cationic CDs of low charge density (CD-PEHA and CD-DMEDA) evoked similar responses in the mono-cultures, whereas in the co-culture, the three cationic CDs ranked according to their charge density (CD-PEI600 > CD-PEHA > CD-DMEDA), when taking into account their inflammatory effect. Thus, the co-culture system developed in this study appears to be a sensitive model for finely discriminating the toxicological profile of cationic nanoparticles differing by the density of their surface charges.
Collapse
Affiliation(s)
| | | | | | | | | | - Carole Ronzani
- Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, UMR 7199, CNRS-Université de Strasbourg, 67400 Illkirch, France; (Y.A.); (J.C.); (M.R.); (L.L.); (F.P.)
| |
Collapse
|
21
|
Bioprinted Multi-Cell Type Lung Model for the Study of Viral Inhibitors. Viruses 2021; 13:v13081590. [PMID: 34452455 PMCID: PMC8402746 DOI: 10.3390/v13081590] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022] Open
Abstract
Influenza A virus (IAV) continuously causes epidemics and claims numerous lives every year. The available treatment options are insufficient and the limited pertinence of animal models for human IAV infections is hampering the development of new therapeutics. Bioprinted tissue models support studying pathogenic mechanisms and pathogen-host interactions in a human micro tissue environment. Here, we describe a human lung model, which consisted of a bioprinted base of primary human lung fibroblasts together with monocytic THP-1 cells, on top of which alveolar epithelial A549 cells were printed. Cells were embedded in a hydrogel consisting of alginate, gelatin and collagen. These constructs were kept in long-term culture for 35 days and their viability, expression of specific cell markers and general rheological parameters were analyzed. When the models were challenged with a combination of the bacterial toxins LPS and ATP, a release of the proinflammatory cytokines IL-1β and IL-8 was observed, confirming that the model can generate an immune response. In virus inhibition assays with the bioprinted lung model, the replication of a seasonal IAV strain was restricted by treatment with an antiviral agent in a dose-dependent manner. The printed lung construct provides an alveolar model to investigate pulmonary pathogenic biology and to support development of new therapeutics not only for IAV, but also for other viruses.
Collapse
|
22
|
Goldsteen PA, Yoseif C, Dolga AM, Gosens R. Human pluripotent stem cells for the modelling and treatment of respiratory diseases. Eur Respir Rev 2021; 30:30/161/210042. [PMID: 34348980 PMCID: PMC9488746 DOI: 10.1183/16000617.0042-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
Respiratory diseases are among the leading causes of morbidity and mortality worldwide, representing a major unmet medical need. New chemical entities rarely make it into the clinic to treat respiratory diseases, which is partially due to a lack of adequate predictive disease models and the limited availability of human lung tissues to model respiratory disease. Human pluripotent stem cells (hPSCs) may help fill this gap by serving as a scalable human in vitro model. In addition, human in vitro models of rare genetic mutations can be generated using hPSCs. hPSC-derived epithelial cells and organoids have already shown great potential for the understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments. These potentials can also be applied to other hPSC-derived lung cell types in the future. In this review, we will discuss how hPSCs have brought, and may continue to bring, major changes to the field of respiratory diseases by understanding the molecular mechanisms of the pathology and by finding efficient therapeutics. Human pluripotent stem cells may help to develop animal-free, fully human in vitro models to advance our understanding of disease mechanisms, for finding new potential targets by using high-throughput screening platforms, and for personalised treatments.https://bit.ly/3cahaqz
Collapse
Affiliation(s)
- Pien A Goldsteen
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands .,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Christina Yoseif
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Dept of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Viola H, Washington K, Selva C, Grunwell J, Tirouvanziam R, Takayama S. A High-Throughput Distal Lung Air-Blood Barrier Model Enabled By Density-Driven Underside Epithelium Seeding. Adv Healthc Mater 2021; 10:e2100879. [PMID: 34174173 DOI: 10.1002/adhm.202100879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Indexed: 12/18/2022]
Abstract
High-throughput tissue barrier models can yield critical insights on how barrier function responds to therapeutics, pathogens, and toxins. However, such models often emphasize multiplexing capability at the expense of physiologic relevance. Particularly, the distal lung's air-blood barrier is typically modeled with epithelial cell monoculture, neglecting the substantial contribution of endothelial cell feedback in the coordination of barrier function. An obstacle to establishing high-throughput coculture models relevant to the epithelium/endothelium interface is the requirement for underside cell seeding, which is difficult to miniaturize and automate. Therefore, this paper describes a scalable, low-cost seeding method that eliminates inversion by optimizing medium density to float cells so they attach under the membrane. This method generates a 96-well model of the distal lung epithelium-endothelium barrier with serum-free, glucocorticoid-free air-liquid differentiation. The polarized epithelial-endothelial coculture exhibits mature barrier function, appropriate intercellular junction staining, and epithelial-to-endothelial transmission of inflammatory stimuli such as polyinosine:polycytidylic acid (poly(I:C)). Further, exposure to influenza A virus PR8 and human beta-coronavirus OC43 initiates a dose-dependent inflammatory response that propagates from the epithelium to endothelium. While this model focuses on the air-blood barrier, the underside seeding method is generalizable to various coculture tissue models for scalable, physiologic screening.
Collapse
Affiliation(s)
- Hannah Viola
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology 311 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| | - Kendra Washington
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Cauviya Selva
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
| | - Jocelyn Grunwell
- Division of Critical Care Medicine Children's Healthcare of Atlanta at Egleston 1405 Clifton Road NE Atlanta GA 30322 USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics Emory University School of Medicine and Center for CF & Airways Disease Research 2015 Uppergate Dr NE, Rm 344 Atlanta GA 30322 USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30308 USA
- Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology 315 Ferst Dr. NW Atlanta GA 30332 USA
| |
Collapse
|
24
|
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: 5.3] [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
|
25
|
Adams W, Espicha T, Estipona J. Getting Your Neutrophil: Neutrophil Transepithelial Migration in the Lung. Infect Immun 2021; 89:IAI.00659-20. [PMID: 33526562 DOI: 10.1128/iai.00659-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neutrophil transepithelial migration is a fundamental process that facilitates the rapid trafficking of neutrophils to inflammatory foci and occurs across a diverse range of tissues. For decades there has been widespread interest in understanding the mechanisms that drive this migratory process in response to different pathogens and organ systems. This has led to the successful integration of key findings on neutrophil transepithelial migration from the intestines, lungs, liver, genitourinary tract, and other tissues into a single, cohesive model. However, recent studies have identified organ specific differences in neutrophil transepithelial migration. These findings support a model where the tissue in concert with the pro-inflammatory stimuli dictate a unique collection of signals that drive neutrophil trafficking. This review focuses on the mechanisms that drive neutrophil transepithelial migration in response to microbial infection of a single organ, the lung. Herein we provide a detailed analysis of the adhesion molecules and chemoattractants that contribute to the recruitment of neutrophil into the airways. We also highlight important advances in experimental models for studying neutrophil transepithelial migration in the lung over the last decade.
Collapse
Affiliation(s)
- Walter Adams
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Taylor Espicha
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Janine Estipona
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| |
Collapse
|
26
|
Optimizations of In Vitro Mucus and Cell Culture Models to Better Predict In Vivo Gene Transfer in Pathological Lung Respiratory Airways: Cystic Fibrosis as an Example. Pharmaceutics 2020; 13:pharmaceutics13010047. [PMID: 33396283 PMCID: PMC7823756 DOI: 10.3390/pharmaceutics13010047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 11/17/2022] Open
Abstract
The respiratory epithelium can be affected by many diseases that could be treated using aerosol gene therapy. Among these, cystic fibrosis (CF) is a lethal inherited disease characterized by airways complications, which determine the life expectancy and the effectiveness of aerosolized treatments. Beside evaluations performed under in vivo settings, cell culture models mimicking in vivo pathophysiological conditions can provide complementary insights into the potential of gene transfer strategies. Such models must consider multiple parameters, following the rationale that proper gene transfer evaluations depend on whether they are performed under experimental conditions close to pathophysiological settings. In addition, the mucus layer, which covers the epithelial cells, constitutes a physical barrier for gene delivery, especially in diseases such as CF. Artificial mucus models featuring physical and biological properties similar to CF mucus allow determining the ability of gene transfer systems to effectively reach the underlying epithelium. In this review, we describe mucus and cellular models relevant for CF aerosol gene therapy, with a particular emphasis on mucus rheology. We strongly believe that combining multiple pathophysiological features in single complex cell culture models could help bridge the gaps between in vitro and in vivo settings, as well as viral and non-viral gene delivery strategies.
Collapse
|
27
|
Lan Y, Ng CT, Ong RXS, Muniasamy U, Baeg GH, Ong CN, Yu LE, Bay BH. Urban PM 2.5 reduces angiogenic ability of endothelial cells in an alveolar-capillary co-culture lung model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 202:110932. [PMID: 32800216 DOI: 10.1016/j.ecoenv.2020.110932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 06/11/2023]
Abstract
Adverse health effects arising from exposure to fine particulates have become a major concern. Angiogenesis is a vital physiological process for the growth and development of cells and structures in the human body, whereby excessive or insufficient vessel growth could contribute to pathogenesis of diseases. We therefore evaluated indirect effects of carbon black (CB) and inhalable airborne particles on the angiogenic ability of unexposed Human Umbilical Vein Endothelial Cells (HUVECs) by co-culturing HUVECs with pre-exposed Small Airway Epithelial Cells (SAECs). As endothelial cells are major components of blood vessels and potential targets of fine particles, we investigated if lung epithelial cells exposed to ambient PM2.5 surrogates could induce bystander effects on neighboring unexposed endothelial cells in an alveolar-capillary co-culture lung model. Epithelial exposure to CB at a non-toxic dose of 25 μg/mL reduced endothelial tube formation and cell adhesion in co-cultured HUVECs, and decreased expression of angiogenic genes in SAECs. Similarly, exposure of differentiated SAECs to PM2.5 surrogates reduced cell reproductive ability, adhesion and tube formation of neighboring HUVECs. This indicates epithelial exposure to CB and urban PM2.5 surrogates both compromised the angiogenic ability of endothelial cells through bystander effects, thereby potentially perturbing the ventilation-perfusion ratio and affecting lung function.
Collapse
Affiliation(s)
- Yang Lan
- Department of Civil and Environmental Engineering, 1 Engineering Drive 2, National University of Singapore, Singapore, 117576, Singapore
| | - Cheng Teng Ng
- Department of Anatomy, 4 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; NUS Environmental Research Institute, 5A Engineering Drive 1, National University of Singapore, Singapore
| | - Rui Xin Sherlyn Ong
- Department of Anatomy, 4 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Umamaheswari Muniasamy
- Department of Anatomy, 4 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore; NUS Environmental Research Institute, 5A Engineering Drive 1, National University of Singapore, Singapore
| | - Gyeong Hun Baeg
- Department of Anatomy, 4 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Choon Nam Ong
- NUS Environmental Research Institute, 5A Engineering Drive 1, National University of Singapore, Singapore; Saw Swee Hock School of Public Health, 12 Science Drive 2, National University of Singapore, Singapore, 117549, Singapore
| | - Liya E Yu
- Department of Civil and Environmental Engineering, 1 Engineering Drive 2, National University of Singapore, Singapore, 117576, Singapore; NUS Environmental Research Institute, 5A Engineering Drive 1, National University of Singapore, Singapore
| | - Boon Huat Bay
- Department of Anatomy, 4 Medical Drive, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
28
|
Canivet L, Denayer FO, Dubot P, Garçon G, Lo Guidice JM. Toxicity of iron nanoparticles towards primary cultures of human bronchial epithelial cells. J Appl Toxicol 2020; 41:203-215. [PMID: 32767597 DOI: 10.1002/jat.4033] [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: 04/14/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 11/08/2022]
Abstract
Air pollution is a public health issue and the toxicity of ambient particulate matter (PM) is well-recognized. Although it does not mostly contribute to the total mass of PM, increasing evidence indicates that the ultrafine fraction has generally a greater toxicity than the others do. A better knowledge of the underlying mechanisms involved in the pathological disorders related to nanoparticles (NPs) remains essential. Hence, the goal of this study was to determine better whether the exposure to a relatively low dose of well-characterized iron-rich NPs (Fe-NPs) might alter some critical toxicological endpoints in a relevant primary culture model of human bronchial epithelial cells (HBECs). We sought to use Fe-NPs representative of those frequently found in the industrial smokes of metallurgical industries. After having noticed the effective internalization of Fe-NPs, oxidative, inflammatory, DNA repair, and apoptotic endpoints were investigated within HBECs, mainly through transcriptional screening. Taken together, these results revealed that, despite it only produced relatively low levels of reactive oxygen species without any significant oxidative damage, low-dose Fe-NPs quickly significantly deregulated the transcription of some target genes closely involved in the proinflammatory response. Although this inflammatory process seemed to stay under control over time in case of this acute scenario of exposure, the future study of its evolution after a scenario of repeated exposure could be very interesting to evaluate the toxicity of Fe-NPs better.
Collapse
Affiliation(s)
- Ludivine Canivet
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS-IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France
| | - Franck-Olivier Denayer
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS-IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France
| | - Pierre Dubot
- CNRS UMR 7182, Métaux et céramiques à microstructure contrôlée, Institut de Chimie et des Matériaux, Paris Est, Thiais, France
| | - Guillaume Garçon
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS-IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France
| | - J-M Lo Guidice
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483-IMPECS-IMPact de l'Environnement Chimique sur la Santé, F-59000 Lille, France
| |
Collapse
|
29
|
Development of a miniaturized 96-Transwell air-liquid interface human small airway epithelial model. Sci Rep 2020; 10:13022. [PMID: 32747751 PMCID: PMC7400554 DOI: 10.1038/s41598-020-69948-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
In order to overcome the challenges associated with a limited number of airway epithelial cells that can be obtained from clinical sampling and their restrained capacity to divide ex vivo, miniaturization of respiratory drug discovery assays is of pivotal importance. Thus, a 96-well microplate system was developed where primary human small airway epithelial (hSAE) cells were cultured at an air–liquid interface (ALI). After four weeks of ALI culture, a pseudostratified epithelium containing basal, club, goblet and ciliated cells was produced. The 96-well ALI cultures displayed a cellular composition, ciliary beating frequency, and intercellular tight junctions similar to 24-well conditions. A novel custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements, together with dextran permeability measurements, confirmed that the 96-well culture developed a tight barrier function during ALI differentiation. 96-well hSAE cultures were responsive to transforming growth factor β1 (TGF-β1) and tumor necrosis factor α (TNF-α) in a concentration dependent manner. Thus, the miniaturized cellular model system enables the recapitulation of a physiologically responsive, differentiated small airway epithelium, and a robotic integration provides a medium throughput approach towards pharmaceutical drug discovery, for instance, in respect of fibrotic distal airway/lung diseases.
Collapse
|
30
|
Valacchi G, Magnani N, Woodby B, Ferreira SM, Evelson P. Particulate Matter Induces Tissue OxInflammation: From Mechanism to Damage. Antioxid Redox Signal 2020; 33:308-326. [PMID: 32443938 DOI: 10.1089/ars.2019.8015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Oxidative stress and oxidative damage are central hypothetical mechanisms for the adverse effects of airborne particulate matter (PM). Activation of inflammatory cells capable of generating reactive oxygen and nitrogen species is another proposed damage pathway. Understanding the interplay between these responses can help us understand the adverse health effects attributed to breathing polluted air. Recent Advances: The consequences of PM exposure on different organs are oxidative damage, decreased function, and inflammation, which can lead to the development/exacerbation of proinflammatory disorders. Mitochondrial damage is also an important event in PM-induced cytotoxicity. Critical Issues: Reactive oxygen species (ROS) are generated during phagocytosis of the particles, leading to enhancement of oxidative stress and triggering the inflammatory response. The activation of inflammatory signaling pathways results in the release of cytokines and other mediators, which can further induce ROS production by activating endogenous enzymes, leading to a positive feedback loop, which can aggravate the effects triggered by PM exposure. Future Directions: Further research is required to elucidate the exact mechanisms by which PM exposure results in adverse health effects, in terms of the relationship between the redox responses triggered by the presence of the particles and the inflammation observed in the different organs, so the development/exacerbation of PM-associated health problems can be prevented.
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, North Carolina, USA.,Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy.,Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Brittany Woodby
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, North Carolina, USA
| | - Sandra María Ferreira
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica, Buenos Aires, Argentina.,CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
31
|
Mathieu E, MacPherson CW, Belvis J, Mathieu O, Robert V, Saint-Criq V, Langella P, Tompkins TA, Thomas M. Oral Primo-Colonizing Bacteria Modulate Inflammation and Gene Expression in Bronchial Epithelial Cells. Microorganisms 2020; 8:microorganisms8081094. [PMID: 32707845 PMCID: PMC7464694 DOI: 10.3390/microorganisms8081094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023] Open
Abstract
The microbiota of the mouth disperses into the lungs, and both compartments share similar phyla. Considering the importance of the microbiota in the maturation of the immunity and physiology during the first days of life, we hypothesized that primo-colonizing bacteria of the oral cavity may induce immune responses in bronchial epithelial cells. Herein, we have isolated and characterized 57 strains of the buccal cavity of two human newborns. These strains belong to Streptococcus, Staphylococcus, Enterococcus, Rothia and Pantoea genera, with Streptococcus being the most represented. The strains were co-incubated with a bronchial epithelial cell line (BEAS-2B), and we established their impact on a panel of cytokines/chemokines and global changes in gene expression. The Staphylococcus strains, which appeared soon after birth, induced a high production of IL-8, suggesting they can trigger inflammation, whereas the Streptococcus strains were less associated with inflammation pathways. The genera Streptococcus, Enterococcus and Pantoea induced differential profiles of cytokine/chemokine/growth factor and set of genes associated with maturation of morphology. Altogether, our results demonstrate that the microorganisms, primo-colonizing the oral cavity, impact immunity and morphology of the lung epithelial cells, with specific effects depending on the phylogeny of the strains.
Collapse
Affiliation(s)
- Elliot Mathieu
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (E.M.); (V.R.); (V.S.-C.); (P.L.)
| | - Chad W. MacPherson
- Rosell Institute for Microbiome and Probiotics, Lallemand Health Solutions Inc., Montreal, QC H4P 2R2, Canada; (C.W.M.); (J.B.); (O.M.); (T.A.T.)
| | - Jocelyn Belvis
- Rosell Institute for Microbiome and Probiotics, Lallemand Health Solutions Inc., Montreal, QC H4P 2R2, Canada; (C.W.M.); (J.B.); (O.M.); (T.A.T.)
| | - Olivier Mathieu
- Rosell Institute for Microbiome and Probiotics, Lallemand Health Solutions Inc., Montreal, QC H4P 2R2, Canada; (C.W.M.); (J.B.); (O.M.); (T.A.T.)
| | - Véronique Robert
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (E.M.); (V.R.); (V.S.-C.); (P.L.)
| | - Vinciane Saint-Criq
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (E.M.); (V.R.); (V.S.-C.); (P.L.)
| | - Philippe Langella
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (E.M.); (V.R.); (V.S.-C.); (P.L.)
| | - Thomas A. Tompkins
- Rosell Institute for Microbiome and Probiotics, Lallemand Health Solutions Inc., Montreal, QC H4P 2R2, Canada; (C.W.M.); (J.B.); (O.M.); (T.A.T.)
| | - Muriel Thomas
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350 Jouy-en-Josas, France; (E.M.); (V.R.); (V.S.-C.); (P.L.)
- Correspondence:
| |
Collapse
|
32
|
Rapiteanu R, Karagyozova T, Zimmermann N, Singh K, Wayne G, Martufi M, Belyaev NN, Hessel EM, Michalovich D, Macarron R, Rowan WC, Cairns WJ, Roger J, Betts J, Beinke S, Maratou K. Highly efficient genome editing in primary human bronchial epithelial cells differentiated at air–liquid interface. Eur Respir J 2020; 55:13993003.00950-2019. [DOI: 10.1183/13993003.00950-2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 01/31/2020] [Indexed: 12/25/2022]
|
33
|
Artzy-Schnirman A, Zidan H, Elias-Kirma S, Ben-Porat L, Tenenbaum-Katan J, Carius P, Fishler R, Schneider-Daum N, Lehr CM, Sznitman J. Capturing the Onset of Bacterial Pulmonary Infection in Acini-On-Chips. ADVANCED BIOSYSTEMS 2019; 3:e1900026. [PMID: 32648651 PMCID: PMC7611792 DOI: 10.1002/adbi.201900026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/30/2019] [Indexed: 12/20/2022]
Abstract
Bacterial invasion of the respiratory system leads to complex immune responses. In the deep alveolar regions, the first line of defense includes foremost the alveolar epithelium, the surfactant-rich liquid lining, and alveolar macrophages. Typical in vitro models come short of mimicking the complexity of the airway environment in the onset of airway infection; among others, they neither capture the relevant anatomical features nor the physiological flows innate of the acinar milieu. Here, novel microfluidic-based acini-on-chips that mimic more closely the native acinar airways at a true scale with an anatomically inspired, multigeneration alveolated tree are presented and an inhalation-like maneuver is delivered. Composed of human alveolar epithelial lentivirus immortalized cells and macrophages-like human THP-1 cells at an air-liquid interface, the models maintain critically an epithelial barrier with immune function. To demonstrate, the usability and versatility of the platforms, a realistic inhalation exposure assay mimicking bacterial infection is recapitulated, whereby the alveolar epithelium is exposed to lipopolysaccharides droplets directly aerosolized and the innate immune response is assessed by monitoring the secretion of IL8 cytokines. These efforts underscore the potential to deliver advanced in vitro biosystems that can provide new insights into drug screening as well as acute and subacute toxicity assays.
Collapse
Affiliation(s)
- Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hikaia Zidan
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Shani Elias-Kirma
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lee Ben-Porat
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Janna Tenenbaum-Katan
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Patrick Carius
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany
- Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Ramy Fishler
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nicole Schneider-Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, 66123 Saarbrücken, Germany
- Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
34
|
Ladel S, Schlossbauer P, Flamm J, Luksch H, Mizaikoff B, Schindowski K. Improved In Vitro Model for Intranasal Mucosal Drug Delivery: Primary Olfactory and Respiratory Epithelial Cells Compared with the Permanent Nasal Cell Line RPMI 2650. Pharmaceutics 2019; 11:pharmaceutics11080367. [PMID: 31374872 PMCID: PMC6723747 DOI: 10.3390/pharmaceutics11080367] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 12/28/2022] Open
Abstract
Background: The epithelial layer of the nasal mucosa is the first barrier for drug permeation during intranasal drug delivery. With increasing interest for intranasal pathways, adequate in vitro models are required. Here, porcine olfactory (OEPC) and respiratory (REPC) primary cells were characterised against the nasal tumour cell line RPMI 2650. Methods: Culture conditions for primary cells from porcine nasal mucosa were optimized and the cells characterised via light microscope, RT-PCR and immunofluorescence. Epithelial barrier function was analysed via transepithelial electrical resistance (TEER), and FITC-dextran was used as model substance for transepithelial permeation. Beating cilia necessary for mucociliary clearance were studied by immunoreactivity against acetylated tubulin. Results: OEPC and REPC barrier models differ in TEER, transepithelial permeation and MUC5AC levels. In contrast, RPMI 2650 displayed lower levels of MUC5AC, cilia markers and TEER, and higher FITC-dextran flux rates. Conclusion: To screen pharmaceutical formulations for intranasal delivery in vitro, translational mucosal models are needed. Here, a novel and comprehensive characterisation of OEPC and REPC against RPMI 2650 is presented. The established primary models display an appropriate model for nasal mucosa with secreted MUC5AC, beating cilia and a functional epithelial barrier, which is suitable for long-term evaluation of sustained release dosage forms.
Collapse
Affiliation(s)
- Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Patrick Schlossbauer
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany
| | - Harald Luksch
- School of Life Sciences, Technical University of Munich, Liesel-Beckmann-Straße 4, 85354 Freising-Weihenstephan, Germany
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Hubertus-Liebrecht Straße 35, 88400 Biberach, Germany.
| |
Collapse
|
35
|
Liu G, Betts C, Cunoosamy DM, Åberg PM, Hornberg JJ, Sivars KB, Cohen TS. Use of precision cut lung slices as a translational model for the study of lung biology. Respir Res 2019; 20:162. [PMID: 31324219 PMCID: PMC6642541 DOI: 10.1186/s12931-019-1131-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/09/2019] [Indexed: 12/28/2022] Open
Abstract
Animal models remain invaluable for study of respiratory diseases, however, translation of data generated in genetically homogeneous animals housed in a clean and well-controlled environment does not necessarily provide insight to the human disease situation. In vitro human systems such as air liquid interface (ALI) cultures and organ-on-a-chip models have attempted to bridge the divide between animal models and human patients. However, although 3D in nature, these models struggle to recreate the architecture and complex cellularity of the airways and parenchyma, and therefore cannot mimic the complex cell-cell interactions in the lung. To address this issue, lung slices have emerged as a useful ex vivo tool for studying the respiratory responses to inflammatory stimuli, infection, and novel drug compounds. This review covers the practicality of precision cut lung slice (PCLS) generation and benefits of this ex vivo culture system in modeling human lung biology and disease pathogenesis.
Collapse
Affiliation(s)
- Guanghui Liu
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine Betts
- Pathology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Danen M Cunoosamy
- Bioscience, Respiratory Inflammation and Autoimmunity, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Present Address: Sanofi, Cambridge, MA, USA
| | - Per M Åberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jorrit J Hornberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kinga Balogh Sivars
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Taylor S Cohen
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20877, USA.
| |
Collapse
|
36
|
Lee D, Chambers M. A bilayer tissue culture model of the bovine alveolus. F1000Res 2019; 8:357. [PMID: 31448101 PMCID: PMC6685456 DOI: 10.12688/f1000research.18696.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2019] [Indexed: 08/02/2024] Open
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture bilayer model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and the bilayer cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the bilayer model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their culture as a bilayer in conjunction with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The bilayer model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
37
|
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the co-culture model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their co-culture with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The co-culture model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
38
|
Medium throughput breathing human primary cell alveolus-on-chip model. Sci Rep 2018; 8:14359. [PMID: 30254327 PMCID: PMC6156575 DOI: 10.1038/s41598-018-32523-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Organs-on-chips have the potential to improve drug development efficiency and decrease the need for animal testing. For the successful integration of these devices in research and industry, they must reproduce in vivo contexts as closely as possible and be easy to use. Here, we describe a ‘breathing’ lung-on-chip array equipped with a passive medium exchange mechanism that provide an in vivo-like environment to primary human lung alveolar cells (hAEpCs) and primary lung endothelial cells. This configuration allows the preservation of the phenotype and the function of hAEpCs for several days, the conservation of the epithelial barrier functionality, while enabling simple sampling of the supernatant from the basal chamber. In addition, the chip design increases experimental throughput and enables trans-epithelial electrical resistance measurements using standard equipment. Biological validation revealed that human primary alveolar type I (ATI) and type II-like (ATII) epithelial cells could be successfully cultured on the chip over multiple days. Moreover, the effect of the physiological cyclic strain showed that the epithelial barrier permeability was significantly affected. Long-term co-culture of primary human lung epithelial and endothelial cells demonstrated the potential of the lung-on-chip array for reproducible cell culture under physiological conditions. Thus, this breathing lung-on-chip array, in combination with patients’ primary ATI, ATII, and lung endothelial cells, has the potential to become a valuable tool for lung research, drug discovery and precision medicine.
Collapse
|
39
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
40
|
Tenenbaum-Katan J, Artzy-Schnirman A, Fishler R, Korin N, Sznitman J. Biomimetics of the pulmonary environment in vitro: A microfluidics perspective. BIOMICROFLUIDICS 2018; 12:042209. [PMID: 29887933 PMCID: PMC5973897 DOI: 10.1063/1.5023034] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/20/2018] [Indexed: 05/08/2023]
Abstract
The entire luminal surface of the lungs is populated with a complex yet confluent, uninterrupted airway epithelium in conjunction with an extracellular liquid lining layer that creates the air-liquid interface (ALI), a critical feature of healthy lungs. Motivated by lung disease modelling, cytotoxicity studies, and drug delivery assessments amongst other, in vitro setups have been traditionally conducted using macroscopic cultures of isolated airway cells under submerged conditions or instead using transwell inserts with permeable membranes to model the ALI architecture. Yet, such strategies continue to fall short of delivering a sufficiently realistic physiological in vitro airway environment that cohesively integrates at true-scale three essential pillars: morphological constraints (i.e., airway anatomy), physiological conditions (e.g., respiratory airflows), and biological functionality (e.g., cellular makeup). With the advent of microfluidic lung-on-chips, there have been tremendous efforts towards designing biomimetic airway models of the epithelial barrier, including the ALI, and leveraging such in vitro scaffolds as a gateway for pulmonary disease modelling and drug screening assays. Here, we review in vitro platforms mimicking the pulmonary environment and identify ongoing challenges in reconstituting accurate biological airway barriers that still widely prevent microfluidic systems from delivering mainstream assays for the end-user, as compared to macroscale in vitro cell cultures. We further discuss existing hurdles in scaling up current lung-on-chip designs, from single airway models to more physiologically realistic airway environments that are anticipated to deliver increasingly meaningful whole-organ functions, with an outlook on translational and precision medicine.
Collapse
Affiliation(s)
- Janna Tenenbaum-Katan
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Rami Fishler
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| |
Collapse
|
41
|
Bhowmick R, Derakhshan T, Liang Y, Ritchey J, Liu L, Gappa-Fahlenkamp H. A Three-Dimensional Human Tissue-Engineered Lung Model to Study Influenza A Infection. Tissue Eng Part A 2018; 24:1468-1480. [PMID: 29732955 DOI: 10.1089/ten.tea.2017.0449] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) claims ∼250,000-500,000 lives annually worldwide. Currently, there are a few in vitro models available to study IAV immunopathology. Monolayer cultures of cell lines and primary lung cells (two-dimensional [2D] cell culture) is the most commonly used tool, however, this system does not have the in vivo-like structure of the lung and immune responses to IAV as it lacks the three-dimensional (3D) tissue structure. To recapitulate the lung physiology in vitro, a system that contains multiple cell types within a 3D environment that allows cell movement and interaction would provide a critical tool. In this study, as a first step in designing a 3D-Human Tissue-Engineered Lung Model (3D-HTLM), we describe the 3D culture of primary human small airway epithelial cells (HSAEpCs) and determined the immunophenotype of this system in response to IAV infections. We constructed a 3D chitosan-collagen scaffold and cultured HSAEpCs on these scaffolds at air-liquid interface (ALI). These 3D cultures were compared with 2D-cultured HSAEpCs for viability, morphology, marker protein expression, and cell differentiation. Results showed that the 3D-cultured HSAEpCs at ALI yielded maximum viable cells and morphologically resembled the in vivo lower airway epithelium. There were also significant increases in aquaporin-5 and cytokeratin-14 expression for HSAEpCs cultured in 3D compared to 2D. The 3D culture system was used to study the infection of HSAEpCs with two major IAV strains, H1N1 and H3N2. The HSAEpCs showed distinct changes in marker protein expression, both at mRNA and protein levels, and the release of proinflammatory cytokines. This study is the first step in the development of the 3D-HTLM, which will have wide applicability in studying pulmonary pathophysiology and therapeutics development.
Collapse
Affiliation(s)
- Rudra Bhowmick
- 1 School of Chemical Engineering, Oklahoma State University , Stillwater, Oklahoma
| | - Tahereh Derakhshan
- 1 School of Chemical Engineering, Oklahoma State University , Stillwater, Oklahoma
| | - Yurong Liang
- 2 Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | - Jerry Ritchey
- 3 Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | - Lin Liu
- 2 Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University , Stillwater, Oklahoma
| | | |
Collapse
|
42
|
Zscheppang K, Berg J, Hedtrich S, Verheyen L, Wagner DE, Suttorp N, Hippenstiel S, Hocke AC. Human Pulmonary 3D Models For Translational Research. Biotechnol J 2018; 13:1700341. [PMID: 28865134 PMCID: PMC7161817 DOI: 10.1002/biot.201700341] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/23/2017] [Indexed: 12/13/2022]
Abstract
Lung diseases belong to the major causes of death worldwide. Recent innovative methodological developments now allow more and more for the use of primary human tissue and cells to model such diseases. In this regard, the review covers bronchial air-liquid interface cultures, precision cut lung slices as well as ex vivo cultures of explanted peripheral lung tissue and de-/re-cellularization models. Diseases such as asthma or infections are discussed and an outlook on further areas for development is given. Overall, the progress in ex vivo modeling by using primary human material could make translational research activities more efficient by simultaneously fostering the mechanistic understanding of human lung diseases while reducing animal usage in biomedical research.
Collapse
Affiliation(s)
- Katja Zscheppang
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Johanna Berg
- Department of BiotechnologyTechnical University of BerlinGustav‐Meyer‐Allee 25Berlin 13335Germany
| | - Sarah Hedtrich
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Leonie Verheyen
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Darcy E. Wagner
- Helmholtz Zentrum Munich, Lung Repair and Regeneration Unit, Comprehensive Pneumology CenterMember of the German Center for Lung ResearchMunichGermany
| | - Norbert Suttorp
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Stefan Hippenstiel
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Andreas C. Hocke
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| |
Collapse
|
43
|
Ewart L, Dehne EM, Fabre K, Gibbs S, Hickman J, Hornberg E, Ingelman-Sundberg M, Jang KJ, Jones DR, Lauschke VM, Marx U, Mettetal JT, Pointon A, Williams D, Zimmermann WH, Newham P. Application of Microphysiological Systems to Enhance Safety Assessment in Drug Discovery. Annu Rev Pharmacol Toxicol 2017; 58:65-82. [PMID: 29029591 DOI: 10.1146/annurev-pharmtox-010617-052722] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Enhancing the early detection of new therapies that are likely to carry a safety liability in the context of the intended patient population would provide a major advance in drug discovery. Microphysiological systems (MPS) technology offers an opportunity to support enhanced preclinical to clinical translation through the generation of higher-quality preclinical physiological data. In this review, we highlight this technological opportunity by focusing on key target organs associated with drug safety and metabolism. By focusing on MPS models that have been developed for these organs, alongside other relevant in vitro models, we review the current state of the art and the challenges that still need to be overcome to ensure application of this technology in enhancing drug discovery.
Collapse
Affiliation(s)
- Lorna Ewart
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | | | - Kristin Fabre
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451, USA
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, 1081 LA Amsterdam, The Netherlands
| | - James Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, USA
| | - Ellinor Hornberg
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, 431 83 Mölndal, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - David R Jones
- Medicines & Healthcare Products Regulatory Agency, London SW1W 9SZ, United Kingdom
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Jerome T Mettetal
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451, USA
| | - Amy Pointon
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | - Dominic Williams
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK), Goettingen 37075, Germany
| | - Peter Newham
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| |
Collapse
|
44
|
Bovard D, Iskandar A, Luettich K, Hoeng J, Peitsch MC. Organs-on-a-chip. TOXICOLOGY RESEARCH AND APPLICATION 2017. [DOI: 10.1177/2397847317726351] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the last few years, considerable attention has been given to in vitro models in an attempt to reduce the use of animals and to decrease the rate of preclinical failure associated with the development of new drugs. Simple two-dimensional cultures grown in a dish are now frequently replaced by organotypic cultures with three-dimensional (3-D) architecture, which enables interactions between cells, promoting their differentiation and increasing their in vivo likeness. Microengineering now enables the incorporation of small devices into 3-D culture models to reproduce the complex microenvironment of the modeled organ, often referred to as organs-on-a-chip (OoCs). This review describes various OoCs developed to mimic liver, brain, kidney, and lung tissues. Current challenges encountered in attempts to recreate the in vivo environment are described, as well as some examples of OoCs. Finally, attention is given to the ongoing evolution of OoCs with the aim of solving one of the major limitations in that they can only represent a single organ. Multi-organ-on-a-chip (MOC) systems mimic organ interactions observed in the human body and aim to provide the features of compound uptake, metabolism, and excretion, while simultaneously allowing for insights into biological effects. MOCs might therefore represent a new paradigm in drug development, providing a better understanding of dose responses and mechanisms of toxicity, enabling the detection of drug resistance and supporting the evaluation of pharmacokinetic–pharmacodynamics parameters.
Collapse
Affiliation(s)
- David Bovard
- Philip Morris Products SA, Neuchatel, Switzerland
| | | | | | - Julia Hoeng
- Philip Morris Products SA, Neuchatel, Switzerland
| | | |
Collapse
|
45
|
Leclercq B, Alleman LY, Perdrix E, Riffault V, Happillon M, Strecker A, Lo-Guidice JM, Garçon G, Coddeville P. Particulate metal bioaccessibility in physiological fluids and cell culture media: Toxicological perspectives. ENVIRONMENTAL RESEARCH 2017; 156:148-157. [PMID: 28342961 DOI: 10.1016/j.envres.2017.03.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/21/2017] [Accepted: 03/17/2017] [Indexed: 06/06/2023]
Abstract
According to the literature, tiny amounts of transition metals in airborne fine particles (PM2.5) may induce proinflammatory cell response through reactive oxygen species production. The solubility of particle-bound metals in physiological fluids, i.e. the metal bioaccessibility is driven by factors such as the solution chemical composition, the contact time with the particles, and the solid-to-liquid phase ratio (S/L). In this work, PM2.5-bound metal bioaccessibility was assessed in various physiological-like solutions including cell culture media in order to evidence the potential impact on normal human bronchial epithelial cells (NHBE) when studying the cytotoxicity and inflammatory responses of PM2.5 towards the target bronchial compartment. Different fluids (H2O, PBS, LHC-9 culture medium, Gamble and human respiratory mucus collected from COPD patients), various S/L conditions (from 1/6000 to 1/100,000) and exposure times (6, 24 and 72h) were tested on urban PM2.5 samples. In addition, metals' total, soluble and insoluble fractions from PM2.5 in LHC-9 were deposited on NHBE cells (BEAS-2B) to measure their cytotoxicity and inflammatory potential (i.e., G6PDH activity, secretion of IL-6 and IL-8). The bioaccessibility is solution-dependent. A higher salinity or organic content may increase or inhibit the bioaccessibiliy according to the element, as observed in the complex mucus matrix. Decreasing the S/L ratio also affect the bioaccessibility depending on the solution tested while the exposure time appears less critical. The LHC-9 culture medium appears to be a good physiological proxy as it induces metal bioaccessibilities close to the mucus values and is little affected by S/L ratios or exposure time. Only the insoluble fraction can be linked to the PM2.5-induced cytotoxicity. By contrast, both soluble and insoluble fractions can be related to the secretion of cytokines. The metal bioaccessibility in LHC-9 of the total, soluble, and insoluble fractions of the PM2.5 under study did not explain alone, the cytotoxicity nor the inflammatory response observed in BEAS-2B cells. These findings confirm the urgent need to perform further toxicological studies to better evaluate the synergistic effect of both bioaccessible particle-bound metals and organic species.
Collapse
Affiliation(s)
- Bérénice Leclercq
- IMT Lille Douai, Univ. Lille, SAGE - Département Sciences de l'Atmosphère et Génie de l'Environnement, F-59000 Lille, France; Univ. Lille, CHU Lille, Institut Pasteur de Lille, EA4483-IMPECS, France
| | - Laurent Yves Alleman
- IMT Lille Douai, Univ. Lille, SAGE - Département Sciences de l'Atmosphère et Génie de l'Environnement, F-59000 Lille, France.
| | - Esperanza Perdrix
- IMT Lille Douai, Univ. Lille, SAGE - Département Sciences de l'Atmosphère et Génie de l'Environnement, F-59000 Lille, France
| | - Véronique Riffault
- IMT Lille Douai, Univ. Lille, SAGE - Département Sciences de l'Atmosphère et Génie de l'Environnement, F-59000 Lille, France
| | - Mélanie Happillon
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, EA4483-IMPECS, France
| | | | | | - Guillaume Garçon
- Univ. Lille, CHU Lille, Institut Pasteur de Lille, EA4483-IMPECS, France
| | - Patrice Coddeville
- IMT Lille Douai, Univ. Lille, SAGE - Département Sciences de l'Atmosphère et Génie de l'Environnement, F-59000 Lille, France
| |
Collapse
|
46
|
Rigoni VLS, Kwasniewski FH, Vieira RP, Linhares IS, da Silva JLV, Nogueira-Pedro A, Zamuner SR. Human bronchial epithelial cells injury and cytokine production induced by Tityus serrulatus scorpion venom: An in vitro study. Toxicon 2016; 120:22-8. [PMID: 27452928 DOI: 10.1016/j.toxicon.2016.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/13/2016] [Accepted: 07/20/2016] [Indexed: 12/18/2022]
|