1
|
Jülke EM, Özbay B, Nowicki M, Els-Heindl S, Immig K, Mörl K, Bechmann I, Beck-Sickinger AG. Intranasal Application of Peptides Modulating the Neuropeptide Y System. ACS Pharmacol Transl Sci 2025; 8:1168-1181. [PMID: 40242586 PMCID: PMC11997893 DOI: 10.1021/acsptsci.5c00082] [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: 01/31/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025]
Abstract
The neuropeptide Y multireceptor-multiligand system plays an important role in multiple physiological processes. Targeting the neuropeptide Y1 (Y1R) and Y2 (Y2R) receptors has gained interest in treating weight and mental disorders. Nose-to-brain delivery is an effective tool to overcome the challenges of peptide delivery to cerebral structures. In this study, fluorescently labeled peptides that selectively activate either Y1R or Y2R were studied. The permeability of these compounds was evaluated on Calu-3 cells, a model system of the nasal mucosa. Particular attention was paid to the stability of peptides, and translocation of the intact compounds was demonstrated by combining a permeability assay with a receptor activation assay. Two compounds, selectively targeting either Y1R or Y2R, were selected, and their uptake after intranasal application was analyzed in vivo. Two different imaging systems were compared: whole slide scanning and confocal microscopy. Both methods allow detecting specific signals from the fluorescently labeled peptides. While whole slide scanning provides a comprehensive anatomical overview, confocal microscopy offers an improved signal-to-noise ratio. Finally, peptide-specific signals were quantified over time, displaying rapid peptide uptake within the first 15 min and sustained signals for up to 24 h. Overall, cell-based and in vivo assays were combined to select peptides with high pharmacological potential for nasal applications.
Collapse
Affiliation(s)
- Eva-Maria Jülke
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany
| | - Benginur Özbay
- Institute
of Anatomy, Faculty of Medicine, Leipzig
University, Liebigstraße
13, 04103 Leipzig, Germany
| | - Marcin Nowicki
- Institute
of Anatomy, Faculty of Medicine, Leipzig
University, Liebigstraße
13, 04103 Leipzig, Germany
| | - Sylvia Els-Heindl
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany
| | - Kerstin Immig
- Institute
of Anatomy, Faculty of Medicine, Leipzig
University, Liebigstraße
13, 04103 Leipzig, Germany
| | - Karin Mörl
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany
| | - Ingo Bechmann
- Institute
of Anatomy, Faculty of Medicine, Leipzig
University, Liebigstraße
13, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute
of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, 04103 Leipzig, Germany
| |
Collapse
|
2
|
Chai S, Lee Y, Owens RM, Lee HR, Lee Y, Kim W, Jung S. Dynamic monitoring of a 3D-printed airway tissue model using an organic electrochemical transistor. Biomaterials 2025; 314:122806. [PMID: 39260031 DOI: 10.1016/j.biomaterials.2024.122806] [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: 03/06/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024]
Abstract
Assessing the transepithelial resistance to ion flow in the presence of an electric field enables the evaluation of the integrity of the epithelial cell layer. In this study, we introduce an organic electrochemical transistor (OECT) interfaced with a 3D living tissue, designed to monitor the electrical resistance of cellular barriers in real-time. We have developed a non-invasive, tissue-sensing platform by integrating an inkjet-printed large-area OECT with a 3D-bioprinted multilayered airway tissue. This unique configuration enables the evaluation of epithelial barrier integrity through the dynamic response capabilities of the OECT. Our system effectively tracks the formation and integrity of 3D-printed airway tissues in both liquid-liquid and air-liquid interface culture environments. Furthermore, we successfully quantified the degradation of barrier function due to influenza A (H1N1) viral infection and the dose-dependent efficacy of oseltamivir (Tamiflu®) in mitigating this degradation. The tissue-electronic platform offers a non-invasive and label-free method for real-time monitoring of 3D artificial tissue barriers, without disturbing the cellular biology. It holds the potential for further applications in monitoring the structures and functions of 3D tissues and organs, significantly contributing to the advancement of personalized medicine.
Collapse
Affiliation(s)
- Seungjin Chai
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea
| | - Yunji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge, CB3 0AS, UK
| | - Hwa-Rim Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea
| | - Yongwoo Lee
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea.
| | - Woojo Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea.
| | - Sungjune Jung
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea; Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
3
|
Eltanameli B, Piñeiro-Llanes J, Cristofoletti R. Recent advances in cell-based in vitro models for predicting drug permeability across brain, intestinal, and pulmonary barriers. Expert Opin Drug Metab Toxicol 2024; 20:439-458. [PMID: 38850058 DOI: 10.1080/17425255.2024.2366390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/06/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Recent years have witnessed remarkable progress in the development of cell-based in vitro models aimed at predicting drug permeability, particularly focusing on replicating the barrier properties of the blood-brain barrier (BBB), intestinal epithelium, and lung epithelium. AREA COVERED This review provides an overview of 2D in vitro platforms, including monocultures and co-culture systems, highlighting their respective advantages and limitations. Additionally, it discusses tools and techniques utilized to overcome these limitations, paving the way for more accurate predictions of drug permeability. Furthermore, this review delves into emerging technologies, particularly microphysiological systems (MPS), encompassing static platforms such as organoids and dynamic platforms like microfluidic devices. Literature searches were performed using PubMed and Google Scholar. We focus on key terms such as in vitro permeability models, MPS, organoids, intestine, BBB, and lungs. EXPERT OPINION The potential of these MPS to mimic physiological conditions more closely offers promising avenues for drug permeability assessment. However, transitioning these advanced models from bench to industry requires rigorous validation against regulatory standards. Thus, there is a pressing need to validate MPS to industry and regulatory agency standards to exploit their potential in drug permeability prediction fully. This review underscores the importance of such validation processes to facilitate the translation of these innovative technologies into routine pharmaceutical practice.
Collapse
Affiliation(s)
- Bassma Eltanameli
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Janny Piñeiro-Llanes
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
4
|
Osman N, Curley P, Box H, Liptrott N, Sexton D, Saleem I. In vitro evaluation of physicochemical-dependent effects of polymeric nanoparticles on their cellular uptake and co-localization using pulmonary calu-3 cell lines. Drug Dev Ind Pharm 2024; 50:376-386. [PMID: 38533688 DOI: 10.1080/03639045.2024.2332889] [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: 11/21/2023] [Accepted: 03/15/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE The study evaluated physicochemical properties of eight different polymeric nanoparticles (NPs) and their interaction with lung barrier and their suitability for pulmonary drug delivery. METHODS Eight physiochemically different NPs were fabricated from Poly lactic-co-glycolic acid (PLGA, PL) and Poly glycerol adipate-co-ω-pentadecalactone (PGA-co-PDL, PG) via emulsification-solvent evaporation. Pulmonary barrier integrity was investigated in vitro using Calu-3 under air-liquid interface. NPs internalization was investigated using a group of pharmacological inhibitors with subsequent microscopic visual confirmation. RESULTS Eight NPs were successfully formulated from two polymers using emulsion-solvent evaporation; 200, 500 and 800 nm, negatively-charged and positively-charged. All different NPs did not alter tight junctions and PG NPs showed similar behavior to PL NPs, indicating its suitability for pulmonary drug delivery. Active endocytosis uptake mechanisms with physicochemical dependent manner were observed. In addition, NPs internalization and co-localization with lysosomes were visually confirmed indicating their vesicular transport. CONCLUSION PG and PL NPs had shown no or low harmful effects on the barrier integrity, and with effective internalization and vesicular transport, thus, prospectively can be designed for pulmonary delivery applications.
Collapse
Affiliation(s)
- Nashwa Osman
- Nanoformulations and drug delivery group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- Faculty of Medicine, Sohag University, Egypt
| | - Paul Curley
- Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Helen Box
- Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Neill Liptrott
- Centre of Excellence for Long-acting Therapeutics (CELT), Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
- Immunocompatibility Group, Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Darren Sexton
- Nanoformulations and drug delivery group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Imran Saleem
- Nanoformulations and drug delivery group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
5
|
Meziu E, Shehu K, Koch M, Schneider M, Kraegeloh A. Impact of mucus modulation by N-acetylcysteine on nanoparticle toxicity. Int J Pharm X 2023; 6:100212. [PMID: 37771516 PMCID: PMC10522980 DOI: 10.1016/j.ijpx.2023.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023] Open
Abstract
Human respiratory mucus is a biological hydrogel that forms a protective barrier for the underlying epithelium. Modulation of the mucus layer has been employed as a strategy to enhance transmucosal drug carrier transport. However, a drawback of this strategy is a potential reduction of the mucus barrier properties, in particular in situations with an increased exposure to particles. In this study, we investigated the impact of mucus modulation on its protective role. In vitro mucus was produced by Calu-3 cells, cultivated at the air-liquid interface for 21 days and used for further testing as formed on top of the cells. Analysis of confocal 3D imaging data revealed that after 21 days Calu-3 cells secrete a mucus layer with a thickness of 24 ± 6 μm. Mucus appeared to restrict penetration of 500 nm carboxyl-modified polystyrene particles to the upper 5-10 μm of the layer. Furthermore, a mucus modulation protocol using aerosolized N-acetylcysteine (NAC) was developed. This treatment enhanced the penetration of particles through the mucus down to deeper layers by means of the mucolytic action of NAC. These findings were supported by cytotoxicity data, indicating that intact mucus protects the underlying epithelium from particle-induced effects on membrane integrity. The impact of NAC treatment on the protective properties of mucus was probed by using 50 and 100 nm amine-modified and 50 nm carboxyl-modified polystyrene nanoparticles, respectively. Cytotoxicity was only induced by the amine-modified particles in combination with NAC treatment, implying a reduced protective function of modulated mucus. Overall, our data emphasize the importance of integrating an assessment of the protective function of mucus into the development of therapy approaches involving mucus modulation.
Collapse
Affiliation(s)
- Enkeleda Meziu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Kristela Shehu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Annette Kraegeloh
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| |
Collapse
|
6
|
Silva S, Bicker J, Falcão A, Fortuna A. Air-liquid interface (ALI) impact on different respiratory cell cultures. Eur J Pharm Biopharm 2023; 184:62-82. [PMID: 36696943 DOI: 10.1016/j.ejpb.2023.01.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/24/2022] [Accepted: 01/19/2023] [Indexed: 01/23/2023]
Abstract
The intranasal route has been receiving greater attention from the scientific community not only for systemic drug delivery but also for the treatment of pulmonary and neurological diseases. Along with it, drug transport and permeability studies across the nasal mucosa have exponentially increased. Nevertheless, the translation of data from in vitro cell lines to in vivo studies is not always reliable, due to the difficulty in generating an in vitro model that resembles respiratory human physiology. Among all currently available methodologies, the air-liquid interface (ALI) method is advantageous to promote cell differentiation and optimize the morphological and histological characteristics of airway epithelium cells. Cells grown under ALI conditions, in alternative to submerged conditions, appear to provide relevant input for inhalation and pulmonary toxicology and complement in vivo experiments. Different methodologies and a variety of materials have been used to induce ALI conditions in primary cells and numerous cell lines. Until this day, with only exploratory results, no consensus has been reached regarding the validation of the ALI method, hampering data comparison. The present review describes the most adequate cell models of airway epithelium and how these models are differently affected by ALI conditions. It includes the evaluation of cellular features before and after ALI, and the application of the method in primary cell cultures, commercial 3D primary cells, cell lines and stem-cell derived models. A variety of these models have been recently applied for pharmacological studies against severe acute respiratory syndrome-coronavirus(-2) SARS-CoV(-2), namely primary cultures with alveolar type II epithelium cells and organotypic 3D models. The herein compiled data suggest that ALI conditions must be optimized bearing in mind the type of cells (nasal, bronchial, alveolar), their origin and the objective of the study.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
7
|
Building three-dimensional lung models for studying pharmacokinetics of inhaled drugs. Adv Drug Deliv Rev 2021; 170:386-395. [PMID: 32971227 DOI: 10.1016/j.addr.2020.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/15/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022]
Abstract
Drug development is a critical step in the development pipeline of pharmaceutical industry, commonly performed in traditional cell culture and animal models. Though, those models hold critical gapsin the prediction and the translation of human pharmacokinetic (PK) and pharmacodynamics (PD) parameters. The advances in tissue engineering have allowed the combination of cell biology with microengineering techniques, offering alternatives to conventional preclinical models. Organ-on-a-chips and three-dimensional (3D) bioprinting models present the potentialityof simulating the physiological and pathological microenvironment of living organs and tissues, constituting this way,more realistic models for the assessment of absorption, distribution, metabolism and excretion (ADME) of drugs. Therefore, this review will focus on lung-on-a-chip and 3D bioprinting techniques for developing lung models that can be usedfor predicting PK/PD parameters.
Collapse
|
8
|
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: 13] [Impact Index Per Article: 2.6] [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
|
9
|
Aguiar JA, Huff RD, Tse W, Stämpfli MR, McConkey BJ, Doxey AC, Hirota JA. Transcriptomic and barrier responses of human airway epithelial cells exposed to cannabis smoke. Physiol Rep 2020; 7:e14249. [PMID: 31646766 PMCID: PMC6811686 DOI: 10.14814/phy2.14249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/25/2019] [Accepted: 09/04/2019] [Indexed: 01/04/2023] Open
Abstract
Globally, many jurisdictions are legalizing or decriminalizing cannabis, creating a potential public health issue that would benefit from experimental evidence to inform policy, government regulations, and user practices. Tobacco smoke exposure science has created a body of knowledge that demonstrates the conclusive negative impacts on respiratory health; similar knowledge remains to be established for cannabis. To address this unmet need, we performed in vitro functional and transcriptomic experiments with a human airway epithelial cell line (Calu-3) exposed to cannabis smoke, with tobacco smoke as a positive control. Demonstrating the validity of our in vitro model, tobacco smoke induced gene expression profiles that were significantly correlated with gene expression profiles from published tobacco exposure datasets from bronchial brushings and primary human airway epithelial cell cultures. Applying our model to cannabis smoke, we demonstrate that cannabis smoke induced functional and transcriptional responses that overlapped with tobacco smoke. Ontology and pathway analysis revealed that cannabis smoke induced DNA replication and oxidative stress responses. Functionally, cannabis smoke impaired epithelial cell barrier function, antiviral responses, and increased inflammatory mediator production. Our study reveals striking similarities between cannabis and tobacco smoke exposure on impairing barrier function, suppressing antiviral pathways, potentiating of pro-inflammatory mediators, and inducing oncogenic and oxidative stress gene expression signatures. Collectively our data suggest that cannabis smoke exposure is not innocuous and may possess many of the deleterious properties of tobacco smoke, warranting additional studies to support public policy, government regulations, and user practices.
Collapse
Affiliation(s)
- Jennifer A Aguiar
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Ryan D Huff
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wayne Tse
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin R Stämpfli
- Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario
| | - Brendan J McConkey
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada.,Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario
| | - Andrew C Doxey
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada.,Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario
| | - Jeremy A Hirota
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Firestone Institute for Respiratory Health - Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario
| |
Collapse
|
10
|
Yang X, Chen X, Lei T, Qin L, Zhou Y, Hu C, Liu Q, Gao H. The construction of in vitro nasal cavity-mimic M-cell model, design of M cell-targeting nanoparticles and evaluation of mucosal vaccination by nasal administration. Acta Pharm Sin B 2020; 10:1094-1105. [PMID: 32642415 PMCID: PMC7332807 DOI: 10.1016/j.apsb.2020.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/13/2020] [Accepted: 02/16/2020] [Indexed: 12/18/2022] Open
Abstract
In order to better evaluate the transport effect of nanoparticles through the nasal mucosa, an in vitro nasal cavity-mimic model was designed based on M cells. The differentiation of M cells was induced by co-culture of Calu-3 and Raji cells in invert model. The ZO-1 protein staining and the transport of fluorescein sodium and dexamethasone showed that the inverted co-culture model formed a dense monolayer and possessed the transport ability. The differentiation of M cells was observed by up-regulated expression of Sialyl Lewis A antigen (SLAA) and integrin β1, and down-regulated activity of alkaline phosphatase. After targeting M cells with iRGD peptide (cRGDKGPDC), the transport of nanoparticles increased. In vivo, the co-administration of iRGD could result in the increase of nanoparticles transported to the brain through the nasal cavity after intranasal administration. In the evaluation of immune effect in vivo, the nasal administration of OVA-PLGA/iRGD led to more release of IgG, IFN-γ, IL-2 and secretory IgA (sIgA) compared with OVA@PLGA group. Collectively, the study constructed in vitro M cell model, and proved the enhanced effect of targeting towards M cell with iRGD on improving nasal immunity.
Collapse
Affiliation(s)
- Xiaotong Yang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Xianchun Chen
- College of Materials Science and Engineering, Sichuan University, Chengdu 610064, China
| | - Ting Lei
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Lin Qin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Yang Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Chuan Hu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Qingfeng Liu
- Shanghai University of Medicine & Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai 201499, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
In vitro model for predicting acute inhalation toxicity by using a Calu-3 epithelium cytotoxicity assay. J Pharmacol Toxicol Methods 2019; 98:106576. [PMID: 31026561 DOI: 10.1016/j.vascn.2019.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 03/06/2019] [Accepted: 04/20/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION As the current methods to predict the inhalation toxicity of chemicals using animal models are limited, alternative methods are required. We present a new in vitro prediction method for acute inhalation toxicity using the Calu-3 epithelial cytotoxicity assay applicable for water-soluble inhalable chemicals. METHOD To confirm the characteristics of the optimal Calu-3 epithelium, tight-junction formation, morphology, and mucus secretion were verified using scanning electron microscopy, transepithelial electrical resistance analysis, and immunofluorescence after growth in an air-liquid interface (ALI). Sixty chemicals, including 38 positive and 22 negative for acute inhalation toxicity, were selected from the European Chemical Agency chemical database. The cell viability of the exposed cells was assessed using an MTT assay to predict the acute inhalation toxicity by calculating the area under the receiver operating characteristic (ROC) curve and accuracy. RESULTS When cultivated in an ALI, the epithelium was thicker and secreted more mucin than that under submerged cultivation, characteristic of the in vivo respiratory epithelium. The areas under the ROC curve were 0.75 and 0.78 when exposed to chemicals at concentrations of 2.5 and 5%, respectively. The highest accuracy of the methods was 68 and 78% at cut-off values of 85 and 40% cell viability, respectively. DISCUSSION The in vitro model was moderately accurate with good prediction. It is replicable because of its advantages, i.e., the use of cultured cells and the simplicity of the method. Overall, the Calu-3 epithelial cytotoxicity assay may be a useful and simple approach to identify substances that cause acute inhalation toxicity.
Collapse
|
12
|
Effect of the pulmonary deposition and in vitro permeability on the prediction of plasma levels of inhaled budesonide formulation. Int J Pharm 2017; 532:337-344. [DOI: 10.1016/j.ijpharm.2017.08.124] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/31/2022]
|
13
|
Berthelsen R, Byrialsen JP, Holm R, Jacobsen J, Abrahamsson B, Saabye L, Madelung P, Müllertz A. Development of a μDissolution-Permeation model with in situ drug concentration monitoring. J Drug Deliv Sci Technol 2016. [DOI: 10.1016/j.jddst.2016.06.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Rabiatul AR, Lokanathan Y, Rohaina CM, Chowdhury SR, Aminuddin BS, Ruszymah BHI. Surface modification of electrospun poly(methyl methacrylate) (PMMA) nanofibers for the development of in vitro respiratory epithelium model. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:1297-311. [PMID: 26335265 DOI: 10.1080/09205063.2015.1088183] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Scaffold design is an important aspect of in vitro model development. In this study, nanoscaffold surface modification, namely UV radiation and genipin cross-linking to immobilize collagen on the surface of electrospun poly (methyl methacrylate) (PMMA) nanofiber sheet was investigated. Samples were divided into four groups; PMMA nanofibers (PMMA), collagen-coated PMMA nanofibers (PMMACOL), genipin cross-linked collagen-coated PMMA nanofibers (PMMAGEN), and UV-irradiated collagen-coated PMMA nanofibers (PMMAUV). 6 h of UV radiation significantly reduced the hydrophobicity of PMMA nanofibers from (131.88° ± 1.33°) to (110.04° ± 0.27°) (p < 0.05). The amount of collagen immobilized was significantly higher in PMMAGEN group (239.36 ± 16.63 μg collagen/mg nanofibers) (p < 0.05) compared to the other groups. RECs on all scaffold expressed epithelial cell-specific markers (CK18 and CK14), mucin-producing cell marker (MUC5Ac) and were actively proliferating, based on the positive expression of Ki67. Total number of attached cells was significantly the highest in PMMAUV group on day 9 (6.44 × 10(4) ± 2.77 × 10(4) cells/cm(2)) and it has the highest proliferation rate from day 4 to 9 (0.005 ± 0.003 h(-1)) compared to the other groups. Even though PMMAGEN group showed the highest collagen adsorption, in terms of cells attachment and proliferation, PMMAUV group showed a better outcome compared to the other groups. Thus, PMMAUV scaffold is more suitable to be used in the construction of in vitro respiratory epithelial model.
Collapse
Affiliation(s)
- A R Rabiatul
- a Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - Y Lokanathan
- a Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - C M Rohaina
- a Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - S R Chowdhury
- a Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| | - B S Aminuddin
- b Ear, Nose and Throat Consultant Clinic, Ampang Puteri Specialist Hospital , Ampang , Selangor , Malaysia
| | - B H I Ruszymah
- a Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia.,c Faculty of Medicine, Department of Physiology , Universiti Kebangsaan Malaysia , Kuala Lumpur , Malaysia
| |
Collapse
|
15
|
Meindl C, Stranzinger S, Dzidic N, Salar-Behzadi S, Mohr S, Zimmer A, Fröhlich E. Permeation of Therapeutic Drugs in Different Formulations across the Airway Epithelium In Vitro. PLoS One 2015; 10:e0135690. [PMID: 26274590 PMCID: PMC4537286 DOI: 10.1371/journal.pone.0135690] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/26/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pulmonary drug delivery is characterized by short onset times of the effects and an increased therapeutic ratio compared to oral drug delivery. This delivery route can be used for local as well as for systemic absorption applying drugs as single substance or as a fixed dose combination. Drugs can be delivered as nebulized aerosols or as dry powders. A screening system able to mimic delivery by the different devices might help to assess the drug effect in the different formulations and to identify potential interference between drugs in fixed dose combinations. The present study evaluates manual devices used in animal studies for their suitability for cellular studies. METHODS Calu-3 cells were cultured submersed and in air-liquid interface culture and characterized regarding mucus production and transepithelial electrical resistance. The influence of pore size and material of the transwell membranes and of the duration of air-liquid interface culture was assessed. Compounds were applied in solution and as aerosols generated by MicroSprayer IA-1C Aerosolizer or by DP-4 Dry Powder Insufflator using fluorescein and rhodamine 123 as model compounds. Budesonide and formoterol, singly and in combination, served as examples for drugs relevant in pulmonary delivery. RESULTS AND CONCLUSIONS Membrane material and duration of air-liquid interface culture had no marked effect on mucus production and tightness of the cell monolayer. Co-application of budesonide and formoterol, applied in solution or as aerosol, increased permeation of formoterol across cells in air-liquid interface culture. Problems with the DP-4 Dry Powder Insufflator included compound-specific delivery rates and influence on the tightness of the cell monolayer. These problems were not encountered with the MicroSprayer IA-1C Aerosolizer. The combination of Calu-3 cells and manual aerosol generation devices appears suitable to identify interactions of drugs in fixed drug combination products on permeation.
Collapse
Affiliation(s)
- Claudia Meindl
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | | | - Neira Dzidic
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria
| | | | - Stefan Mohr
- Research Center Pharmaceutical Engineering GmbH, Graz, Austria
| | - Andreas Zimmer
- Department of Pharmaceutical Technology, Institute of Pharmaceutical Sciences, Karl-Franzens-University of Graz, Graz, Austria
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| |
Collapse
|
16
|
Paget V, Dekali S, Kortulewski T, Grall R, Gamez C, Blazy K, Aguerre-Chariol O, Chevillard S, Braun A, Rat P, Lacroix G. Specific uptake and genotoxicity induced by polystyrene nanobeads with distinct surface chemistry on human lung epithelial cells and macrophages. PLoS One 2015; 10:e0123297. [PMID: 25875304 PMCID: PMC4398494 DOI: 10.1371/journal.pone.0123297] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/17/2015] [Indexed: 01/10/2023] Open
Abstract
Nanoparticle surface chemistry is known to play a crucial role in interactions with cells and their related cytotoxic effects. As inhalation is a major route of exposure to nanoparticles, we studied specific uptake and damages of well-characterized fluorescent 50 nm polystyrene (PS) nanobeads harboring different functionalized surfaces (non-functionalized, carboxylated and aminated) on pulmonary epithelial cells and macrophages (Calu-3 and THP-1 cell lines respectively). Cytotoxicity of in mass dye-labeled functionalized PS nanobeads was assessed by xCELLigence system and alamarBlue viability assay. Nanobeads-cells interactions were studied by video-microscopy, flow cytometry and also confocal microscopy. Finally ROS generation was assessed by glutathione depletion dosages and genotoxicity was assessed by γ-H2Ax foci detection, which is considered as the most sensitive technique for studying DNA double strand breaks. The uptake kinetic was different for each cell line. All nanobeads were partly adsorbed and internalized, then released by Calu-3 cells, while THP-1 macrophages quickly incorporated all nanobeads which were located in the cytoplasm rather than in the nuclei. In parallel, the genotoxicity study reported that only aminated nanobeads significantly increased DNA damages in association with a strong depletion of reduced glutathione in both cell lines. We showed that for similar nanoparticle concentrations and sizes, aminated polystyrene nanobeads were more cytotoxic and genotoxic than unmodified and carboxylated ones on both cell lines. Interestingly, aminated polystyrene nanobeads induced similar cytotoxic and genotoxic effects on Calu-3 epithelial cells and THP-1 macrophages, for all levels of intracellular nanoparticles tested. Our results strongly support the primordial role of nanoparticles surface chemistry on cellular uptake and related biological effects. Moreover our data clearly show that nanoparticle internalization and observed adverse effects are not necessarily associated.
Collapse
Affiliation(s)
- Vincent Paget
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
| | - Samir Dekali
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC) / UMR CNRS 8638, Faculté de Pharmacie de Paris, Université Paris Descartes (PRES Sorbonne Paris Cité), Paris, France
| | | | - Romain Grall
- CEA, DSV, iRCM, Laboratoire Cancérologie Expérimentale (LCE), Fontenay-aux-Roses, France
| | - Christelle Gamez
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
| | - Kelly Blazy
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
| | - Olivier Aguerre-Chariol
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
| | - Sylvie Chevillard
- CEA, DSV, iRCM, Laboratoire Cancérologie Expérimentale (LCE), Fontenay-aux-Roses, France
| | - Anne Braun
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
| | - Patrice Rat
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC) / UMR CNRS 8638, Faculté de Pharmacie de Paris, Université Paris Descartes (PRES Sorbonne Paris Cité), Paris, France
| | - Ghislaine Lacroix
- Institut National de l'Environnement Industriel et des Risques (INERIS), Unité de Toxicologie Expérimentale, Parc Technologique ALATA, BP2, Verneuil-en-Halatte, France
- * E-mail:
| |
Collapse
|
17
|
Bol L, Galas JC, Hillaireau H, Le Potier I, Nicolas V, Haghiri-Gosnet AM, Fattal E, Taverna M. A microdevice for parallelized pulmonary permeability studies. Biomed Microdevices 2014; 16:277-85. [PMID: 24337430 DOI: 10.1007/s10544-013-9831-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We describe a compartmentalized microdevice specifically designed to perform permeability studies across a model of lung barrier. Epithelial cell barriers were reproduced by culturing Calu-3 cells at the air-liquid interface (AIC) in 1 mm² microwells made from a perforated glass slide with an embedded porous membrane. We created a single basolateral reservoir for all microwells which eliminated the need to renew the growth medium during the culture growth phase. To perform drug permeability studies on confluent cell layers, the cell culture slide was aligned and joined to a collection platform consisting in 35 μL collection reservoirs connected at the top and bottom with microchannels. The integrity and functionality of the cell barriers were demonstrated by measurement of trans-epithelial electrical resistance (TEER), confocal imaging and permeability assays of ¹⁴C-sucrose. Micro-cell barriers were able to form confluent layers in 1 week, demonstrating a similar bioelectrical evolution as the Transwell systems used as controls. Tight junctions were observed throughout the cell-cell interfaces, and the low permeability coefficients of ¹⁴C-sucrose confirmed their functional presence, creating a primary barrier to the diffusion of solutes. This microdevice could facilitate the monitoring of biomolecule transport and the screening of formulations promoting their passage across the pulmonary barrier, in order to select candidates for pulmonary administration to patients.
Collapse
Affiliation(s)
- Ludivine Bol
- Faculté de Pharmacie, Université Paris-Sud, 92290, Châtenay-Malabry, France
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Intranasal chitosan-DNA vaccines that protect across influenza virus subtypes. Int J Pharm 2014; 473:113-25. [DOI: 10.1016/j.ijpharm.2014.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 06/03/2014] [Accepted: 07/02/2014] [Indexed: 12/19/2022]
|
19
|
Dekali S, Gamez C, Kortulewski T, Blazy K, Rat P, Lacroix G. Assessment of an in vitro model of pulmonary barrier to study the translocation of nanoparticles. Toxicol Rep 2014; 1:157-171. [PMID: 28962236 PMCID: PMC5598380 DOI: 10.1016/j.toxrep.2014.03.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 03/10/2014] [Accepted: 03/10/2014] [Indexed: 12/22/2022] Open
Abstract
As the lung is one of the main routes of exposure to manufactured nanoparticles, we developed an in vitro model resembling the alveolo-capillary barrier for the study of nanoparticle translocation. In order to provide a relevant and ethical in vitro model, cost effective and easy-to-implement human cell lines were used. Pulmonary epithelial cells (Calu-3 cell line) and macrophages (THP-1 differentiated cells) were cultivated on the apical side and pulmonary endothelial cells (HPMEC-ST1.6R cell line) on the basal side of a microporous polyester membrane (Transwell®). Translocation of non-functionalized (51 and 110 nm) and aminated (52 nm) fluorescent polystyrene (PS) nanobeads was studied in this system. The use of Calu-3 cells allowed high transepithelial electrical resistance (TEER) values (>1000 Ω cm2) in co-cultures with or without macrophages. After 24 h of exposure to non-cytotoxic concentrations of non-functionalized PS nanobeads, the relative TEER values (%/t0) were significantly decreased in co-cultures. Epithelial cells and macrophages were able to internalize PS nanobeads. Regarding translocation, Transwell® membranes per se limit the passage of nanoparticles between apical and basal side. However, small non-functionalized PS nanobeads (51 nm) were able to translocate as they were detected in the basal side of co-cultures. Altogether, these results show that this co-culture model present good barrier properties allowing the study of nanoparticle translocation but research effort need to be done to improve the neutrality of the porous membrane delimitating apical and basal sides of the model.
Collapse
Affiliation(s)
- Samir Dekali
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes (PRES Sorbonne Paris Cité), 75270 Paris Cedex 06, France
| | - Christelle Gamez
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
| | - Thierry Kortulewski
- CEA, DSV, iRCM, Plateforme imagerie photonique, 92260 Fontenay-aux-Roses, France
| | - Kelly Blazy
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
| | - Patrice Rat
- Laboratoire de chimie et toxicologie analytique et cellulaire (C-TAC), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes (PRES Sorbonne Paris Cité), 75270 Paris Cedex 06, France
| | - Ghislaine Lacroix
- INERIS (Institut National de l’Environnement industriel et des RISques), Unité de Toxicologie expérimentale, 60550 Verneuil-en-Halatte, France
- Corresponding author at: INERIS, Parc technologique ALATA, BP2, 60550, Verneuil-en-Halatte, France. Tel.: +33 3 44 55 63 15; fax: +33 3 44 55 66 05
| |
Collapse
|
20
|
Nøhr MK, Holm R, Thale ZI, Nielsen CU. Intestinal absorption of the antiepileptic drug substance vigabatrin in Göttingen mini-pigs is unaffected by co-administration of amino acids. Int J Pharm 2014; 466:18-20. [DOI: 10.1016/j.ijpharm.2014.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/28/2014] [Accepted: 03/01/2014] [Indexed: 01/21/2023]
|
21
|
Abstract
Abstract Many epithelial cells form polarized monolayers under in vivo and in vitro conditions. Typically, epithelial cells are cultured for differentiation on insert systems where cells are plated on a porous filter membrane. Although the cultured monolayers have been a standard system to study epithelial physiology, there are some limits: The epithelial cells growing inside the commercial inserts are not optimal to visualize directly through lenses on inverted microscopes. The cell images are optically distorted and background fluorescence is bright due to the filter membrane positioned between the cells and the lens. In addition, the cells are not easily accessible by electrodes due to the presence of tall side walls. Here, we present the design, fabrication, and practical applications of an improved system for analysis of polarized epithelial monolayers. This new system allows (1) direct imaging of cells without an interfering filter membrane, (2) electrophysiological measurements, and (3) detection of apical secretion with minimal dilution. Therefore, our culture method is optimized to study differentiated epithelial cells at the single-cell and subcellular levels, and can be extended to other cell types with minor modifications.
Collapse
Affiliation(s)
- Jong Bae Seo
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | | | | |
Collapse
|
22
|
Salomon JJ, Muchitsch VE, Gausterer JC, Schwagerus E, Huwer H, Daum N, Lehr CM, Ehrhardt C. The Cell Line NCl-H441 Is a Useful in Vitro Model for Transport Studies of Human Distal Lung Epithelial Barrier. Mol Pharm 2014; 11:995-1006. [DOI: 10.1021/mp4006535] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Johanna J. Salomon
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Viktoria E. Muchitsch
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Julia C. Gausterer
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Elena Schwagerus
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Hanno Huwer
- Department
of Cardiothoracic Surgery, Völklingen Heart Centre, Völklingen D-66333, Germany
| | - Nicole Daum
- Helmholtz Institute
for Pharmaceutical Research Saarland, Saarbrücken D-66123, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute
for Pharmaceutical Research Saarland, Saarbrücken D-66123, Germany
| | - Carsten Ehrhardt
- School
of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
23
|
Mechanisms of absorption and elimination of drugs administered by inhalation. Ther Deliv 2013; 4:1027-45. [PMID: 23919477 DOI: 10.4155/tde.13.67] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary drug delivery is an effective route for local or systemic drug administration. However, compared with other routes of administration, there is a scarcity of information on how drugs are absorbed from the lung. The different cell composition lining the airways and alveoli makes this task extremely complicated. Lung cell lines and primary culture cells are useful in studying the absorption mechanisms. However, it is imperative that these cell cultures express essential features required to study these mechanisms such as intact tight junctions and transporters. In vivo, the drug has to face defensive physical and immunological barriers such as mucociliary clearance and alveolar macrophages. Knowledge of the physicochemical properties of the drug and aerosol formulation is required. All of these factors interact together leading to either successful drug deposition followed by absorption or drug elimination. These aspects concerning drug transport in the lung are addressed in this review.
Collapse
|
24
|
Ong HX, Traini D, Young PM. Pharmaceutical applications of the Calu-3 lung epithelia cell line. Expert Opin Drug Deliv 2013; 10:1287-302. [PMID: 23730924 DOI: 10.1517/17425247.2013.805743] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION The Calu-3 lung cell line has been shown to be a promising in vitro model of airway epithelia due to its similarity to in vivo physiology. Hence, over the past decade, it has found increasing applications in the pharmaceutical industry. AREAS COVERED This review focuses on the pharmaceutical applications of the Calu-3 cell line in areas such as mechanisms of drug transport, studying aerosol deposition, controlled release studies and identification of possible drug-drug interactions. The main findings of various studies, as well as the predictive potential of this model, are presented and discussed in this review. EXPERT OPINION There is still a lack of mechanistic knowledge regarding transport of inhaled therapeutics across the lungs. Cell culture models such as Calu-3 provide a simple and reproducible system to study the underlying mechanisms by which inhaled therapeutics interact with the lungs. However, more complex systems that integrate particle deposition onto different cell culture systems may be useful in addressing some fundamental questions to generate a better understanding of determinants that influences pulmonary drug dissolution, absorption, metabolism and efficacy. Ultimately the use of the Calu-3 cell line provides a basic research tool that enables the development of safer and more effective inhaled therapeutics.
Collapse
Affiliation(s)
- Hui Xin Ong
- Woolcock Institute of Medical Research, Respiratory Technology, Glebe, NSW, Australia
| | | | | |
Collapse
|
25
|
Mamlouk M, Young PM, Bebawy M, Haghi M, Mamlouk S, Mulay V, Traini D. Salbutamol sulfate absorption across Calu-3 bronchial epithelia cell monolayer is inhibited in the presence of common anionic NSAIDs. J Asthma 2013; 50:334-41. [PMID: 23406450 DOI: 10.3109/02770903.2013.773518] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE The aim of this study was to characterize the permeability kinetics of salbutamol sulfate, a commonly used β2-agonist in the treatment of asthma exacerbation, across Calu-3 respiratory epithelial cell monolayers in the presence of non-steroidal anti-inflammatory drugs (NSAIDs), as they have been implicated to be able to modulate organic cation transporters (OCTs). METHODS Calu-3 cell monolayers were grown in a liquid covered culture (LCC) configuration on 0.33 cm(2) Transwell polyester cell culture supports. Monolayers, cultured between 11 and 14 days were evaluated for epithelial resistance, tight junction integrity, and expression of OCT using Western blot analysis. The transport of salbutamol across the monolayer was studied as a function of concentration. Directional transport was investigated by assessing apical-basal (a-b) and basal-apical (b-a) directions. The influence of a non-specific OCT inhibitor (tetraethylammonium, TEA) and three NSAIDs (aspirin, ibuprofen, and indomethacin) on the uptake of salbutamol was studied. RESULTS The flux of salbutamol sulfate increased with increasing concentration before reaching a plateau, suggesting the involvement of a transport-mediated uptake mechanism. Western blot analysis detected the presence of OCT1-3 and N1 and N2 sub-types, suggesting the presence of functioning transporters. The apparent permeability (P(app)) of 0.1 mM salbutamol across the epithelial monolayer displayed directional transport in the a-b direction which was inhibited by ˜70% in the presence of TEA, suggesting OCT-mediated uptake. Likewise, the uptake of 0.1 mM salbutamol was decreased in the presence of all the three NSAIDs, supporting a mechanism whereby NSAIDs inhibit absorption of salbutamol across the bronchial epithelium via effects on the OCT transporters. CONCLUSION This study demonstrates that NSAIDs influence the uptake kinetics of salbutamol in an in vitro Calu-3 cell system.
Collapse
Affiliation(s)
- Mariam Mamlouk
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
26
|
Boegh M, Foged C, Müllertz A, Mørck Nielsen H. Mucosal drug delivery: barriers, in vitro models and formulation strategies. J Drug Deliv Sci Technol 2013. [DOI: 10.1016/s1773-2247(13)50055-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Benediktsdóttir BE, Arason AJ, Halldórsson S, Gudjónsson T, Másson M, Baldursson Ó. Drug Delivery Characteristics of the Progenitor Bronchial Epithelial Cell Line VA10. Pharm Res 2012; 30:781-91. [DOI: 10.1007/s11095-012-0919-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 10/18/2012] [Indexed: 11/28/2022]
|
28
|
Sarmento B, Andrade F, da Silva SB, Rodrigues F, das Neves J, Ferreira D. Cell-based in vitro models for predicting drug permeability. Expert Opin Drug Metab Toxicol 2012; 8:607-21. [PMID: 22424145 DOI: 10.1517/17425255.2012.673586] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION In vitro cell models have been used to predict drug permeation in early stages of drug development, since they represent an easy and reproducible method, allowing the tracking of drug absorption rate and mechanism, with an advantageous cost-benefit ratio. Such cell-based models are mainly composed of immortalized cells with an intrinsic ability to grow in a monolayer when seeded in permeable supports, maintaining their physiologic characteristics regarding epithelium cell physiology and functionality. AREAS COVERED This review summarizes the most important intestinal, pulmonary, nasal, vaginal, rectal, ocular and skin cell-based in vitro models for predicting the permeability of drugs. Moreover, the similitude between in vitro cell models and in vivo conditions are discussed, providing evidence that each model may provisionally resemble different drug absorption route. EXPERT OPINION Despite the widespread use of in vitro cell models for drug permeability and absorption evaluation purposes, a detailed study on the properties of these models and their in vitro-in vivo correlation compared with human data are required to further use in order to consider a future drug discovery optimization and clinical development.
Collapse
Affiliation(s)
- Bruno Sarmento
- Department of Pharmaceutical Technology, LTF/CICF, Faculty of Pharmacy, University of Porto, Portugal.
| | | | | | | | | | | |
Collapse
|