1
|
Iriondo C, Koornneef S, Skarp KP, Buscop-van Kempen M, Boerema-de Munck A, Rottier RJ. Simple-Flow: A 3D-Printed Multiwell Flow Plate to Coculture Primary Human Lung Cells at the Air-Liquid Interface. ACS Biomater Sci Eng 2025; 11:451-462. [PMID: 39719361 PMCID: PMC11734690 DOI: 10.1021/acsbiomaterials.4c01322] [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: 07/17/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024]
Abstract
Immortalized epithelial cell lines and animal models have been used in fundamental and preclinical research to study pulmonary diseases. However valuable, though, these models incompletely recapitulate the in vivo human lung, which leads to low predictive outcomes in potential respiratory treatments. Advanced technology and cell culture techniques stimulate the development of improved models that more closely mimic the physiology of the human lung. Nonetheless, most of these models are technically demanding and have a low throughput and reproducibility. Here, we describe a robust fluidic device consisting of a biocompatible and customizable 3D-printed cell culture plate, the Simple-Flow, which has medium throughput, is simple to manufacture, and is easy to set up. As a proof of principle, human primary bronchial epithelial cells (hPBECs) and human pulmonary microvascular endothelial cells (hMVECs) were cocultured on the apical and basolateral sides of the inset membranes, respectively. While hPBECs were cultured at the air-liquid interface to induce mucociliary differentiation, hMVECs were exposed to flow medium for up to 2 weeks. We show the versatility of 3D-printing technology in designing in vitro models for cell culturing applications, such as pediatric lung diseases or other pulmonary disorders.
Collapse
Affiliation(s)
- Cinta Iriondo
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Sem Koornneef
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Kari-Pekka Skarp
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Marjon Buscop-van Kempen
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Anne Boerema-de Munck
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Robbert J. Rottier
- Department
of Pediatric Surgery, Sophia Children’s Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department
of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| |
Collapse
|
2
|
Richter C, Latta L, Harig D, Carius P, Stucki JD, Hobi N, Hugi A, Schumacher P, Krebs T, Gamrekeli A, Stöckle F, Urbschat K, Montalvo G, Lautenschläger F, Loretz B, Hidalgo A, Schneider‐Daum N, Lehr C. A stretchable human lung-on-chip model of alveolar inflammation for evaluating anti-inflammatory drug response. Bioeng Transl Med 2025; 10:e10715. [PMID: 39801748 PMCID: PMC11711225 DOI: 10.1002/btm2.10715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/18/2024] [Accepted: 08/03/2024] [Indexed: 01/16/2025] Open
Abstract
This study describes a complex human in vitro model for evaluating anti-inflammatory drug response in the alveoli that may contribute to the reduction of animal testing in the pre-clinical stage of drug development. The model is based on the human alveolar epithelial cell line Arlo co-cultured with macrophages differentiated from the THP-1 cell line, creating a physiological biological microenvironment. To mimic the three-dimensional architecture and dynamic expansion and relaxation of the air-blood-barrier, they are grown on a stretchable microphysiological lung-on-chip. For validating the in vitro model, three different protocols have been developed to demonstrate the clinically established anti-inflammatory effect of glucocorticoids to reduce certain inflammatory markers after different pro-inflammatory stimuli: (1) an inflammation caused by bacterial LPS (lipopolysaccharides) to simulate an LPS-induced acute lung injury measured best with cytokine IL-6 release; (2) an inflammation caused by LPS at ALI (air-liquid interface) to investigate aerosolized anti-inflammatory treatment, measured with chemokine IL-8 release; and (3) an inflammation with a combination of human inflammatory cytokines TNFα and IFNγ to simulate a critical cytokine storm leading to epithelial barrier disruption, where the eventual weakening or protection of the epithelial barrier can be measured. In all cases, the presence of macrophages appeared to be crucial to mediating inflammatory changes in the alveolar epithelium. LPS induction led to inflammatory changes independently of stretch conditions. Dynamic stretch, emulating breathing-like mechanics, was essential for in vitro modeling of the clinically relevant outcome of epithelial barrier disruption upon TNFα/IFNγ-induced inflammation.
Collapse
Affiliation(s)
- Clémentine Richter
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- Department of PharmacySaarland UniversitySaarbrückenGermany
| | - Lorenz Latta
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
| | - Daria Harig
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- Department of PharmacySaarland UniversitySaarbrückenGermany
| | - Patrick Carius
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- Department of PharmacySaarland UniversitySaarbrückenGermany
| | - Janick D. Stucki
- AlveoliX AG, Swiss Organs‐on‐Chip InnovationBernSwitzerland
- ARTORG Center for Biomedical Engineering Research, Organs‐on‐Chip Technologies, University of BernBernSwitzerland
| | - Nina Hobi
- AlveoliX AG, Swiss Organs‐on‐Chip InnovationBernSwitzerland
- ARTORG Center for Biomedical Engineering Research, Organs‐on‐Chip Technologies, University of BernBernSwitzerland
| | - Andreas Hugi
- AlveoliX AG, Swiss Organs‐on‐Chip InnovationBernSwitzerland
| | | | | | | | - Felix Stöckle
- Center for Thorax Medicine, Clinic SaarbrückenSaarbrückenGermany
| | - Klaus Urbschat
- Section of Thoracic Surgery of the Saar Lung Center, SHG ClinicsVölklingenGermany
| | - Galia Montalvo
- Department of Experimental PhysicsSaarland UniversitySaarbrückenGermany
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), School of Medicine, Saarland UniversityHomburgGermany
| | - Franziska Lautenschläger
- Department of Experimental PhysicsSaarland UniversitySaarbrückenGermany
- Center for Biophysics, Saarland UniversitySaarbrückenGermany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
| | - Alberto Hidalgo
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
| | | | - Claus‐Michael Lehr
- Helmholtz Institute for Pharmaceutical Research SaarlandSaarbrückenGermany
- Department of PharmacySaarland UniversitySaarbrückenGermany
| |
Collapse
|
3
|
Koceva H, Mosig A. Human-Induced Pluripotent Stem Cell-Based Alveolus-on-Chip Model to Study Influenza Virus A Infection. Methods Mol Biol 2025; 2890:225-235. [PMID: 39890730 DOI: 10.1007/978-1-0716-4326-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
This chapter introduces an alveolus-on-chip model for studying influenza virus A infection. The model represents a physiologically relevant alternative to animal models and a scalable and cost-effective approach to gaining mechanistic insights into human-related viral infection processes in vitro. We provide a comprehensive protocol for creating a human autologous model from human-induced pluripotent stem cell (hiPSC)-derived alveolar type II cells, endothelial cells, and macrophages. The protocol includes details on on-chip cell culture, microfluidic perfusion, and monitoring influenza A virus infection through immunofluorescence imaging.
Collapse
Affiliation(s)
- Hristina Koceva
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Alexander Mosig
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.
| |
Collapse
|
4
|
Zhukova OA, Ozerskaya IV, Basmanov DV, Stolyarov VY, Bogush VG, Kolesov VV, Zykov KA, Yusubalieva GM, Baklaushev VP. “Lung-on-a-chip” as an instrument for studying the pathophysiology of human respiration. КЛИНИЧЕСКАЯ ПРАКТИКА 2024; 15:70-88. [DOI: 10.17816/clinpract637140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
“Lung-on-a-chip” (LoC) is a microfluidic device, imitating the gas-fluid interface of the pulmonary alveole in the human lung and intended for pathophysiological, pharmacological and molecular-biological studies of the air-blood barrier in vitro. The LoC device itself contains a system of fluid and gas microchannels, separated with a semipermeable elastic membrane, containing a polymer base and the alveolar cell elements. Depending on the type of LoC (single-, double- and three-channel), the membrane may contain only alveolocytes or alveolocytes combined with other cells — endotheliocytes, fibroblasts, alveolar macrophages or tumor cells. Some LoC models also include proteinic or hydrogel stroma, imitating the pulmonary interstitium. The first double-channel LoC variant, in which one side of the membrane contained an alveolocytic monolayer and the other side — a monolayer of endotheliocytes, was developed in 2010 by a group of scientists from the Harvard University for maximally precise in vitro reproduction of the micro-environment and biomechanics operations of the alveoli. Modern LoC modifications include the same elements and differ only by the construction of the microfluidic system, by the biomaterial of semipermeable membrane, by the composition of cellular and stromal elements and by specific tasks to be solved. Besides the LoC imitating the hematoalveolar barrier, there are modifications for studying the specific pathophysiological processes, for the screening of medicinal products, for modeling specific diseases, for example, lung cancer, chronic obstructive pulmonary disease or asthma. In the present review, we have analyzed the existing types of LoC, the biomaterials used, the methods of detecting molecular processes within the microfluidic devices and the main directions of research to be conducted using the “lung-on-a-chip”.
Collapse
Affiliation(s)
- Oksana A. Zhukova
- Pulmonology Scientific Research Institute
- Federal Center of Brain Research and Neurotechnologies
| | | | - Dmitry V. Basmanov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine
| | | | | | | | - Kirill A. Zykov
- Pulmonology Scientific Research Institute
- Russian University of Medicine
| | - Gaukhar M. Yusubalieva
- Federal Center of Brain Research and Neurotechnologies
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies
- Engelhardt Institute of Molecular Biology
| | - Vladimir P. Baklaushev
- Pulmonology Scientific Research Institute
- Federal Center of Brain Research and Neurotechnologies
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies
- Engelhardt Institute of Molecular Biology
| |
Collapse
|
5
|
Kumar A, Vaiphei KK, Gulbake A. A nanotechnology driven effectual localized lung cancer targeting approaches using tyrosine kinases inhibitors: Recent progress, preclinical assessment, challenges, and future perspectives. Int J Pharm 2024; 666:124745. [PMID: 39321904 DOI: 10.1016/j.ijpharm.2024.124745] [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: 07/09/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
The higher incidence and mortality rate among all populations worldwide explains the unmet solutions in the treatment of lung cancer. The evolution of targeted therapies using tyrosine kinase inhibitors (TKI) has encouraged anticancer therapies. However, on-target and off-target effects and the development of drug resistance limited the anticancer potential of such targeted biologics. The advances in nanotechnology-driven-TKI embedded carriers that offered a new path toward lung cancer treatment. It is the inhalation route of administration known for its specific, precise, and efficient drug delivery to the lungs. The development of numerous TKI-nanocarriers through inhalation is proof of TKI growth. The future scopes involve using potential lung cancer biomarkers to achieve localized active cancer-targeting strategies. The adequate knowledge of in vitro absorption models usually helps establish better in vitro - in vivo correlation/extrapolation (IVIVC/E) to successfully evaluate inhalable drugs and drug products. The advanced in vitro and ex vivo lung tissue/ organ models offered better tumor heterogeneity, etiology, and microenvironment heterogeneity. The involvement of lung cancer organoids (LCOs), human organ chip models, and genetically modified mouse models (GEMMs) has resolved the challenges associated with conventional in vitro and in vivo models. To access potential inhalation-based drugtherapies, biological barriers, drug delivery, device-based challenges, and regulatory challenges must be encountered associated with their development. A proper understanding of material toxicity, size-based particle deposition at active disease sites, mucociliary clearance, phagocytosis, and the presence of enzymes and surfactants are required to achieve successful inhalational drug delivery (IDD). This article summarizes the future of lung cancer therapy using targeted drug-mediated inhalation using TKI.
Collapse
Affiliation(s)
- Ankaj Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Klaudi K Vaiphei
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India
| | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Guwahati, Assam 781101, India.
| |
Collapse
|
6
|
Funk MC, Nawroth J, Lehmann M. A breath of the future: a novel human model for COPD and beyond. Eur Respir J 2024; 64:2401531. [PMID: 39638366 DOI: 10.1183/13993003.01531-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Maja C Funk
- Institute of Lung Health and Immunity, Comprehensive Pneumology Center Munich, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Janna Nawroth
- Helmholtz Pioneer Campus and Institute of Biological and Medical Imaging, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Mareike Lehmann
- Institute of Lung Health and Immunity, Comprehensive Pneumology Center Munich, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Institute for Lung Health, Gießen, German Center for Lung Research (DZL), Gießen, Germany
| |
Collapse
|
7
|
Grassi L, Crabbé A. Recreating chronic respiratory infections in vitro using physiologically relevant models. Eur Respir Rev 2024; 33:240062. [PMID: 39142711 PMCID: PMC11322828 DOI: 10.1183/16000617.0062-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/18/2024] [Indexed: 08/16/2024] Open
Abstract
Despite the need for effective treatments against chronic respiratory infections (often caused by pathogenic biofilms), only a few new antimicrobials have been introduced to the market in recent decades. Although different factors impede the successful advancement of antimicrobial candidates from the bench to the clinic, a major driver is the use of poorly predictive model systems in preclinical research. To bridge this translational gap, significant efforts have been made to develop physiologically relevant models capable of recapitulating the key aspects of the airway microenvironment that are known to influence infection dynamics and antimicrobial activity in vivo In this review, we provide an overview of state-of-the-art cell culture platforms and ex vivo models that have been used to model chronic (biofilm-associated) airway infections, including air-liquid interfaces, three-dimensional cultures obtained with rotating-wall vessel bioreactors, lung-on-a-chips and ex vivo pig lungs. Our focus is on highlighting the advantages of these infection models over standard (abiotic) biofilm methods by describing studies that have benefited from these platforms to investigate chronic bacterial infections and explore novel antibiofilm strategies. Furthermore, we discuss the challenges that still need to be overcome to ensure the widespread application of in vivo-like infection models in antimicrobial drug development, suggesting possible directions for future research. Bearing in mind that no single model is able to faithfully capture the full complexity of the (infected) airways, we emphasise the importance of informed model selection in order to generate clinically relevant experimental data.
Collapse
Affiliation(s)
- Lucia Grassi
- Laboratory of Pharmaceutical Microbiology, Ghent University, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Belgium
| |
Collapse
|
8
|
Fallert L, Urigoitia-Asua A, Cipitria A, Jimenez de Aberasturi D. Dynamic 3D in vitro lung models: applications of inorganic nanoparticles for model development and characterization. NANOSCALE 2024; 16:10880-10900. [PMID: 38787741 DOI: 10.1039/d3nr06672j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Being a vital organ exposed to the external environment, the lung is susceptible to a plethora of pathogens and pollutants. This is reflected in high incidences of chronic respiratory diseases, which remain a leading cause of mortality world-wide and pose a persistent global burden. It is thus of paramount importance to improve our understanding of these pathologies and provide better therapeutic options. This necessitates the development of representative and physiologically relevant in vitro models. Advances in bioengineering have enabled the development of sophisticated models that not only capture the three-dimensional architecture of the cellular environment but also incorporate the dynamics of local biophysical stimuli. However, such complex models also require novel approaches that provide reliable characterization. Within this review we explore how 3D bioprinting and nanoparticles can serve as multifaceted tools to develop such dynamic 4D printed in vitro lung models and facilitate their characterization in the context of pulmonary fibrosis and breast cancer lung metastasis.
Collapse
Affiliation(s)
- Laura Fallert
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
| | - Ane Urigoitia-Asua
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Department of Applied Chemistry, University of the Basque Country, 20018 Donostia-San Sebastián, Spain
- POLYMAT, Basque Centre for Macromolecular Design and Engineering, 20018 Donostia-San Sebastián, Spain
| | - Amaia Cipitria
- Group of Bioengineering in Regeneration and Cancer, Biogipuzkoa Health Research Institute, 20014 Donostia-San Sebastián, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Dorleta Jimenez de Aberasturi
- Department of Hybrid Biofunctional Materials, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo de Miramon 194, 20014 Donostia-San Sebastián, Spain.
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), 20014 Donostia-San Sebastián, Spain
| |
Collapse
|
9
|
Sun L, Chen H, Xu D, Liu R, Zhao Y. Developing organs-on-chips for biomedical applications. SMART MEDICINE 2024; 3:e20240009. [PMID: 39188702 PMCID: PMC11236011 DOI: 10.1002/smmd.20240009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 08/28/2024]
Abstract
In recent years, organs-on-chips have been arousing great interest for their bionic and stable construction of crucial human organs in vitro. Compared with traditional animal models and two-dimensional cell models, organs-on-chips could not only overcome the limitations of species difference and poor predict ability but also be capable of reappearing the complex cell-cell interaction, tissue interface, biofluid and other physiological conditions of humans. Therefore, organs-on-chips have been regarded as promising and powerful tools in diverse fields such as biology, chemistry, medicine and so on. In this perspective, we present a review of organs-on-chips for biomedical applications. After introducing the key elements and manufacturing craft of organs-on-chips, we intend to review their cut-edging applications in biomedical fields, incorporating biological analysis, drug development, robotics and so on. Finally, the emphasis is focused on the perspectives of organs-on-chips.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
- Mechanobiology InstituteNational University of SingaporeSingaporeSingapore
| | - Hanxu Chen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Dongyu Xu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
10
|
Li L, Bo W, Wang G, Juan X, Xue H, Zhang H. Progress and application of lung-on-a-chip for lung cancer. Front Bioeng Biotechnol 2024; 12:1378299. [PMID: 38854856 PMCID: PMC11157020 DOI: 10.3389/fbioe.2024.1378299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/08/2024] [Indexed: 06/11/2024] Open
Abstract
Lung cancer is a malignant tumour with the highest incidence and mortality worldwide. Clinically effective therapy strategies are underutilized owing to the lack of efficient models for evaluating drug response. One of the main reasons for failure of anticancer drug therapy is development of drug resistance. Anticancer drugs face severe challenges such as poor biodistribution, restricted solubility, inadequate absorption, and drug accumulation. In recent years, "organ-on-a-chip" platforms, which can directly regulate the microenvironment of biomechanics, biochemistry and pathophysiology, have been developed rapidly and have shown great potential in clinical drug research. Lung-on-a-chip (LOC) is a new 3D model of bionic lungs with physiological functions created by micromachining technology on microfluidic chips. This approach may be able to partially replace animal and 2D cell culture models. To overcome drug resistance, LOC realizes personalized prediction of drug response by simulating the lung-related microenvironment in vitro, significantly enhancing therapeutic effectiveness, bioavailability, and pharmacokinetics while minimizing side effects. In this review, we present an overview of recent advances in the preparation of LOC and contrast it with earlier in vitro models. Finally, we describe recent advances in LOC. The combination of this technology with nanomedicine will provide an accurate and reliable treatment for preclinical evaluation.
Collapse
Affiliation(s)
- Lantao Li
- Department of Anesthesiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Guangyan Wang
- Department of General Internal Medicine, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xin Juan
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Haiyi Xue
- Department of Intensive Care Unit, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hongwei Zhang
- Department of Anesthesiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Wang Q, Yang Y, Chen Z, Li B, Niu Y, Li X. Lymph Node-on-Chip Technology: Cutting-Edge Advances in Immune Microenvironment Simulation. Pharmaceutics 2024; 16:666. [PMID: 38794327 PMCID: PMC11124897 DOI: 10.3390/pharmaceutics16050666] [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: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-a-chip technology is attracting growing interest across various domains as a crucial platform for drug screening and testing and is set to play a significant role in precision medicine research. Lymph nodes, being intricately structured organs essential for the body's adaptive immune responses to antigens and foreign particles, are pivotal in assessing the immunotoxicity of novel pharmaceuticals. Significant progress has been made in research on the structure and function of the lymphatic system. However, there is still an urgent need to develop prospective tools and techniques to delve deeper into its role in various diseases' pathological and physiological processes and to develop corresponding immunotherapeutic therapies. Organ chips can accurately reproduce the specific functional areas in lymph nodes to better simulate the complex microstructure of lymph nodes and the interactions between different immune cells, which is convenient for studying specific biological processes. This paper reviews existing lymph node chips and their design approaches. It discusses the applications of the above systems in modeling immune cell motility, cell-cell interactions, vaccine responses, drug testing, and cancer research. Finally, we summarize the challenges that current research faces in terms of structure, cell source, and extracellular matrix simulation of lymph nodes, and we provide an outlook on the future direction of integrated immune system chips.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoqiong Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Q.W.); (Y.Y.); (Z.C.); (B.L.); (Y.N.)
| |
Collapse
|
12
|
Vo Q, Benam KH. Advancements in preclinical human-relevant modeling of pulmonary vasculature on-chip. Eur J Pharm Sci 2024; 195:106709. [PMID: 38246431 PMCID: PMC10939731 DOI: 10.1016/j.ejps.2024.106709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/05/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Preclinical human-relevant modeling of organ-specific vasculature offers a unique opportunity to recreate pathophysiological intercellular, tissue-tissue, and cell-matrix interactions for a broad range of applications. Lung vasculature is particularly important due to its involvement in genesis and progression of rare, debilitating disorders as well as common chronic pathologies. Here, we provide an overview of the latest advances in the development of pulmonary vascular (PV) models using emerging microfluidic tissue engineering technology Organs-on-Chips (so-called PV-Chips). We first review the currently reported PV-Chip systems and their key features, and then critically discuss their major limitations in reproducing in vivo-seen and disease-relevant cellularity, localization, and microstructure. We conclude by presenting latest efforts to overcome such technical and biological limitations and future directions.
Collapse
Affiliation(s)
- Quoc Vo
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kambez H Benam
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
13
|
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
|
14
|
Hickey DJ, Golestanian R, Vilfan A. Nonreciprocal interactions give rise to fast cilium synchronization in finite systems. Proc Natl Acad Sci U S A 2023; 120:e2307279120. [PMID: 37756336 PMCID: PMC10556628 DOI: 10.1073/pnas.2307279120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023] Open
Abstract
Motile cilia beat in an asymmetric fashion in order to propel the surrounding fluid. When many cilia are located on a surface, their beating can synchronize such that their phases form metachronal waves. Here, we computationally study a model where each cilium is represented as a spherical particle, moving along a tilted trajectory with a position-dependent active driving force and a position-dependent internal drag coefficient. The model thus takes into account all the essential broken symmetries of the ciliary beat. We show that taking into account the near-field hydrodynamic interactions, the effective coupling between cilia even over an entire beating cycle can become nonreciprocal: The phase of a cilium is more strongly affected by an adjacent cilium on one side than by a cilium at the same distance in the opposite direction. As a result, synchronization starts from a seed at the edge of a group of cilia and propagates rapidly across the system, leading to a synchronization time that scales proportionally to the linear dimension of the system. We show that a ciliary carpet is characterized by three different velocities: the velocity of fluid transport, the phase velocity of metachronal waves, and the group velocity of order propagation. Unlike in systems with reciprocal coupling, boundary effects are not detrimental for synchronization, but rather enable the formation of the initial seed.
Collapse
Affiliation(s)
- David J. Hickey
- Department of Living Matter Physics, Max Planck Institute for Dynamics and Self-Organization, 37077Göttingen, Germany
| | - Ramin Golestanian
- Department of Living Matter Physics, Max Planck Institute for Dynamics and Self-Organization, 37077Göttingen, Germany
- Rudolf Peierls Centre for Theoretical Physics, University of Oxford, OxfordOX1 3PU, United Kingdom
| | - Andrej Vilfan
- Department of Living Matter Physics, Max Planck Institute for Dynamics and Self-Organization, 37077Göttingen, Germany
- Department of Condensed Matter Physics, Jožef Stefan Institute, 1000Ljubljana, Slovenia
| |
Collapse
|
15
|
Shrestha J, Paudel KR, Nazari H, Dharwal V, Bazaz SR, Johansen MD, Dua K, Hansbro PM, Warkiani ME. Advanced models for respiratory disease and drug studies. Med Res Rev 2023; 43:1470-1503. [PMID: 37119028 PMCID: PMC10946967 DOI: 10.1002/med.21956] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/02/2023] [Accepted: 03/17/2023] [Indexed: 04/30/2023]
Abstract
The global burden of respiratory diseases is enormous, with many millions of people suffering and dying prematurely every year. The global COVID-19 pandemic witnessed recently, along with increased air pollution and wildfire events, increases the urgency of identifying the most effective therapeutic measures to combat these diseases even further. Despite increasing expenditure and extensive collaborative efforts to identify and develop the most effective and safe treatments, the failure rates of drugs evaluated in human clinical trials are high. To reverse these trends and minimize the cost of drug development, ineffective drug candidates must be eliminated as early as possible by employing new, efficient, and accurate preclinical screening approaches. Animal models have been the mainstay of pulmonary research as they recapitulate the complex physiological processes, Multiorgan interplay, disease phenotypes of disease, and the pharmacokinetic behavior of drugs. Recently, the use of advanced culture technologies such as organoids and lung-on-a-chip models has gained increasing attention because of their potential to reproduce human diseased states and physiology, with clinically relevant responses to drugs and toxins. This review provides an overview of different animal models for studying respiratory diseases and evaluating drugs. We also highlight recent progress in cell culture technologies to advance integrated models and discuss current challenges and present future perspectives.
Collapse
Affiliation(s)
- Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Hojjatollah Nazari
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Vivek Dharwal
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sajad Razavi Bazaz
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Matt D. Johansen
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of HealthUniversity of TechnologySydneyNew South WalesAustralia
- Faculty of Health, Australian Research Centre in Complementary & Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M. Hansbro
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Majid Ebrahimi Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
- Institute for Biomedical Materials and Devices, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| |
Collapse
|
16
|
Nawroth JC, Roth D, van Schadewijk A, Ravi A, Maulana TI, Senger CN, van Riet S, Ninaber DK, de Waal AM, Kraft D, Hiemstra PS, Ryan AL, van der Does AM. Breathing on chip: Dynamic flow and stretch accelerate mucociliary maturation of airway epithelium in vitro. Mater Today Bio 2023; 21:100713. [PMID: 37455819 PMCID: PMC10339259 DOI: 10.1016/j.mtbio.2023.100713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Human lung function is intricately linked to blood flow and breathing cycles, but it remains unknown how these dynamic cues shape human airway epithelial biology. Here we report a state-of-the-art protocol for studying the effects of dynamic medium and airflow as well as stretch on human primary airway epithelial cell differentiation and maturation, including mucociliary clearance, using an organ-on-chip device. Perfused epithelial cell cultures displayed accelerated maturation and polarization of mucociliary clearance, and changes in specific cell-types when compared to traditional (static) culture methods. Additional application of airflow and stretch to the airway chip resulted in an increase in polarization of mucociliary clearance towards the applied flow, reduced baseline secretion of interleukin-8 and other inflammatory proteins, and reduced gene expression of matrix metalloproteinase (MMP) 9, fibronectin, and other extracellular matrix factors. These results indicate that breathing-like mechanical stimuli are important modulators of airway epithelial cell differentiation and maturation and that their fine-tuned application could generate models of specific epithelial pathologies, including mucociliary (dys)function.
Collapse
Affiliation(s)
- Janna C. Nawroth
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Emulate Inc., Boston, MA, USA
- Helmholtz Pioneer Campus and Institute for Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Abilash Ravi
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sander van Riet
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis K. Ninaber
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorothea Kraft
- Helmholtz Pioneer Campus and Institute for Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg, Germany
| | - Pieter S. Hiemstra
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cells and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
17
|
Shinde A, Illath K, Kasiviswanathan U, Nagabooshanam S, Gupta P, Dey K, Chakrabarty P, Nagai M, Rao S, Kar S, Santra TS. Recent Advances of Biosensor-Integrated Organ-on-a-Chip Technologies for Diagnostics and Therapeutics. Anal Chem 2023; 95:3121-3146. [PMID: 36716428 DOI: 10.1021/acs.analchem.2c05036] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Ashwini Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Uvanesh Kasiviswanathan
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Shalini Nagabooshanam
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Koyel Dey
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Pulasta Chakrabarty
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, Toyohashi 441-8580, Japan
| | - Suresh Rao
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| | - Srabani Kar
- Department of Physics, Indian Institute of Science Education and Research (IISER), Tirupati, Andhra Pradesh 517507, India
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, India
| |
Collapse
|
18
|
Weiss DJ. What is the need and why is it time for innovative models for understanding lung repair and regeneration? Front Pharmacol 2023; 14:1130074. [PMID: 36860303 PMCID: PMC9968746 DOI: 10.3389/fphar.2023.1130074] [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: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
Advances in tissue engineering continue at a rapid pace and have provided novel methodologies and insights into normal cell and tissue homeostasis, disease pathogenesis, and new potential therapeutic strategies. The evolution of new techniques has particularly invigorated the field and span a range from novel organ and organoid technologies to increasingly sophisticated imaging modalities. This is particularly relevant for the field of lung biology and diseases as many lung diseases, including chronic obstructive pulmonary disease (COPD) and idiopathic fibrosis (IPF), among others, remain incurable with significant morbidity and mortality. Advances in lung regenerative medicine and engineering also offer new potential avenues for critical illnesses such as the acute respiratory distress syndrome (ARDS) which also continue to have significant morbidity and mortality. In this review, an overview of lung regenerative medicine with focus on current status of both structural and functional repair will be presented. This will serve as a platform for surveying innovative models and techniques for study, highlighting the need and timeliness for these approaches.
Collapse
|
19
|
Sisodia Y, Shah K, Ali Sayyed A, Jain M, Ali SA, Gondaliya P, Kalia K, Tekade RK. Lung-on-chip microdevices to foster pulmonary drug discovery. Biomater Sci 2023; 11:777-790. [PMID: 36537540 DOI: 10.1039/d2bm00951j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Respiratory diseases account for unprecedented mortality owing to a lack of personalized or insufficient therapeutic interventions. Fostering pulmonary research into managing pulmonary threat requires a potential alternative approach that can mimick the in vivo complexities of the human body. The in vitro miniaturized bionic simulation of the lung holds great potential in the quest for a successful therapeutic intervention. This review discusses the emerging roles of lung-on-chip microfluidic simulator devices in fostering translational pulmonary drug discovery and personalized medicine. This review also explicates how the lung-on-chip model emulates the breathing patterns, elasticity, and vascularization of lungs in creating a 3D pulmonary microenvironment.
Collapse
Affiliation(s)
- Yashi Sisodia
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Komal Shah
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Adil Ali Sayyed
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Transplantation, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Meenakshi Jain
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Syed Ansar Ali
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Piyush Gondaliya
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.,Department of Transplantation, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Kiran Kalia
- Department of Biotechnology, National of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India
| | - Rakesh Kumar Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, 382355, India.
| |
Collapse
|
20
|
Fu A, Chang M, Zhu H, Liu H, Wu D, Zeng H. Air-blood barrier (ABB) on a chip. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
21
|
Zommiti M, Connil N, Tahrioui A, Groboillot A, Barbey C, Konto-Ghiorghi Y, Lesouhaitier O, Chevalier S, Feuilloley MGJ. Organs-on-Chips Platforms Are Everywhere: A Zoom on Biomedical Investigation. Bioengineering (Basel) 2022; 9:646. [PMID: 36354557 PMCID: PMC9687856 DOI: 10.3390/bioengineering9110646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 08/28/2023] Open
Abstract
Over the decades, conventional in vitro culture systems and animal models have been used to study physiology, nutrient or drug metabolisms including mechanical and physiopathological aspects. However, there is an urgent need for Integrated Testing Strategies (ITS) and more sophisticated platforms and devices to approach the real complexity of human physiology and provide reliable extrapolations for clinical investigations and personalized medicine. Organ-on-a-chip (OOC), also known as a microphysiological system, is a state-of-the-art microfluidic cell culture technology that sums up cells or tissue-to-tissue interfaces, fluid flows, mechanical cues, and organ-level physiology, and it has been developed to fill the gap between in vitro experimental models and human pathophysiology. The wide range of OOC platforms involves the miniaturization of cell culture systems and enables a variety of novel experimental techniques. These range from modeling the independent effects of biophysical forces on cells to screening novel drugs in multi-organ microphysiological systems, all within microscale devices. As in living biosystems, the development of vascular structure is the salient feature common to almost all organ-on-a-chip platforms. Herein, we provide a snapshot of this fast-evolving sophisticated technology. We will review cutting-edge developments and advances in the OOC realm, discussing current applications in the biomedical field with a detailed description of how this technology has enabled the reconstruction of complex multi-scale and multifunctional matrices and platforms (at the cellular and tissular levels) leading to an acute understanding of the physiopathological features of human ailments and infections in vitro.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| | | | | | | | | | | | | | | | - Marc G. J. Feuilloley
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| |
Collapse
|
22
|
Bezrukov A, Galyametdinov Y. Activation and Switching of Supramolecular Chemical Signals in Multi-Output Microfluidic Devices. MICROMACHINES 2022; 13:1778. [PMID: 36296131 PMCID: PMC9611873 DOI: 10.3390/mi13101778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 05/27/2023]
Abstract
In this study, we report on the developing of a continuous microfluidic reaction device that allows selective activation of polyelectrolyte-surfactant chemical signals in microflows and switches them between multiple outputs. A numerical model was developed for convection-diffusion reaction processes in reactive polymer-colloid microfluidic flows. Matlab scripts and scaling laws were developed for this model to predict reaction initiation and completion conditions in microfluidic devices and the location of the reaction front. The model allows the optimization of microfluidic device geometry and the setting of operation modes that provide release of the reaction product through specific outputs. Representing a chemical signal, polyelectrolyte-surfactant reaction products create various logic gate states at microfluidic chip outputs. Such systems may have potential as biochemical signal transmitters in organ-on-chip applications or chemical logic gates in cascaded microfluidic devices.
Collapse
Affiliation(s)
- Artem Bezrukov
- Department of Physical and Colloid Chemistry, Kazan National Research Technological University, Kazan 420015, Russia
| | | |
Collapse
|
23
|
Sengupta A, Roldan N, Kiener M, Froment L, Raggi G, Imler T, de Maddalena L, Rapet A, May T, Carius P, Schneider-Daum N, Lehr CM, Kruithof-de Julio M, Geiser T, Marti TM, Stucki JD, Hobi N, Guenat OT. A New Immortalized Human Alveolar Epithelial Cell Model to Study Lung Injury and Toxicity on a Breathing Lung-On-Chip System. FRONTIERS IN TOXICOLOGY 2022; 4:840606. [PMID: 35832493 PMCID: PMC9272139 DOI: 10.3389/ftox.2022.840606] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
The evaluation of inhalation toxicity, drug safety and efficacy assessment, as well as the investigation of complex disease pathomechanisms, are increasingly relying on in vitro lung models. This is due to the progressive shift towards human-based systems for more predictive and translational research. While several cellular models are currently available for the upper airways, modelling the distal alveolar region poses several constraints that make the standardization of reliable alveolar in vitro models relatively difficult. In this work, we present a new and reproducible alveolar in vitro model, that combines a human derived immortalized alveolar epithelial cell line (AXiAEC) and organ-on-chip technology mimicking the lung alveolar biophysical environment (AXlung-on-chip). The latter mimics key features of the in vivo alveolar milieu: breathing-like 3D cyclic stretch (10% linear strain, 0.2 Hz frequency) and an ultrathin, porous and elastic membrane. AXiAECs cultured on-chip were characterized for their alveolar epithelial cell markers by gene and protein expression. Cell barrier properties were examined by TER (Transbarrier Electrical Resistance) measurement and tight junction formation. To establish a physiological model for the distal lung, AXiAECs were cultured for long-term at air-liquid interface (ALI) on-chip. To this end, different stages of alveolar damage including inflammation (via exposure to bacterial lipopolysaccharide) and the response to a profibrotic mediator (via exposure to Transforming growth factor β1) were analyzed. In addition, the expression of relevant host cell factors involved in SARS-CoV-2 infection was investigated to evaluate its potential application for COVID-19 studies. This study shows that AXiAECs cultured on the AXlung-on-chip exhibit an enhanced in vivo-like alveolar character which is reflected into: 1) Alveolar type 1 (AT1) and 2 (AT2) cell specific phenotypes, 2) tight barrier formation (with TER above 1,000 Ω cm2) and 3) reproducible long-term preservation of alveolar characteristics in nearly physiological conditions (co-culture, breathing, ALI). To the best of our knowledge, this is the first time that a primary derived alveolar epithelial cell line on-chip representing both AT1 and AT2 characteristics is reported. This distal lung model thereby represents a valuable in vitro tool to study inhalation toxicity, test safety and efficacy of drug compounds and characterization of xenobiotics.
Collapse
Affiliation(s)
- Arunima Sengupta
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Nuria Roldan
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Mirjam Kiener
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, Bern, Switzerland.,Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Laurène Froment
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Giulia Raggi
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Theo Imler
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | | | - Aude Rapet
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | | | - Patrick Carius
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany.,Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany.,Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany.,Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Saarbrücken, Germany
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Janick D Stucki
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Nina Hobi
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland.,Department of Pulmonary Medicine, Inselspital, Bern University Hospital, Bern, Switzerland.,Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
24
|
Baptista D, Moreira Teixeira L, Barata D, Tahmasebi Birgani Z, King J, van Riet S, Pasman T, Poot AA, Stamatialis D, Rottier RJ, Hiemstra PS, Carlier A, van Blitterswijk C, Habibović P, Giselbrecht S, Truckenmüller R. 3D Lung-on-Chip Model Based on Biomimetically Microcurved Culture Membranes. ACS Biomater Sci Eng 2022; 8:2684-2699. [PMID: 35502997 PMCID: PMC9198974 DOI: 10.1021/acsbiomaterials.1c01463] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
A comparatively straightforward
approach to accomplish more physiological
realism in organ-on-a-chip (OoC) models is through substrate geometry.
There is increasing evidence that the strongly, microscale curved
surfaces that epithelial or endothelial cells experience when lining
small body lumens, such as the alveoli or blood vessels, impact their
behavior. However, the most commonly used cell culture substrates
for modeling of these human tissue barriers in OoCs, ion track-etched
porous membranes, provide only
flat surfaces. Here, we propose a more realistic culture environment
for alveolar cells based on biomimetically microcurved track-etched
membranes. They recreate the mainly spherical geometry of the cells’
native microenvironment. In this feasibility study, the membranes
were given the shape of hexagonally arrayed hemispherical microwells
by an innovative combination of three-dimensional (3D) microfilm (thermo)forming
and ion track technology. Integrated in microfluidic chips, they separated
a top from a bottom cell culture chamber. The microcurved membranes
were seeded by infusion with primary human alveolar epithelial cells.
Despite the pronounced topology, the cells fully lined the alveoli-like
microwell structures on the membranes’ top side. The confluent
curved epithelial cell monolayers could be cultured successfully at
the air−liquid interface for 14 days. Similarly, the top and
bottom sides of the microcurved membranes were seeded with cells from
the Calu-3 lung epithelial cell line and human lung microvascular
endothelial cells, respectively. Thereby, the latter lined the interalveolar
septum-like interspace between the microwells in a network-type fashion,
as in the natural counterpart. The coculture was maintained for 11
days. The presented 3D lung-on-a-chip model might set the stage for
other (micro)anatomically inspired membrane-based OoCs in the future.
Collapse
Affiliation(s)
- Danielle Baptista
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Liliana Moreira Teixeira
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.,Department of Developmental BioEngineering, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - David Barata
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands.,Instituto de Medicina Molecular, Faculty of Medicine, University of Lisbon, Avenida Professor Egas Moniz, 1649-028 Lisbon, Portugal
| | - Zeinab Tahmasebi Birgani
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Jasia King
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Thijs Pasman
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - André A Poot
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery/Cell Biology, Erasmus (University) Medical Center Rotterdam - Sophia Children's Hospital, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Clemens van Blitterswijk
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Pamela Habibović
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
25
|
Journey of organ on a chip technology and its role in future healthcare scenario. APPLIED SURFACE SCIENCE ADVANCES 2022; 9. [PMCID: PMC9000345 DOI: 10.1016/j.apsadv.2022.100246] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Organ on a chip refers to microengineered biomimetic system which reflects structural and functional characteristics of human tissue. It involves biomaterial technology, cell biology and engineering combined together in a miniaturized platform. Several models using different organs such as lungs on a chip, liver on a chip, kidney on a chip, heart on a chip, intestine on a chip and skin on a chip have been successfully developed. Food and Drug administration (FDA) has also shown confidence in this technology and has partnered with industries/institutes which are working with this technology. In this review, the concepts and applications of Organ on a chip model in different scientific domains including disease model development, drug screening, toxicology, pathogenesis study, efficacy testing and virology is discussed. It is envisaged that amalgamation of various organs on chip modules into a unified body on chip device is of utmost importance for diagnosis and treatment, especially considering the complications due to the ongoing COVID-19 pandemic. It is expected that the market demand for developing organ on chip devices to skyrocket in the near future.
Collapse
|
26
|
Three-dimensional models of the lung: past, present and future: a mini review. Biochem Soc Trans 2022; 50:1045-1056. [PMID: 35411381 DOI: 10.1042/bst20190569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/15/2022] [Accepted: 03/04/2022] [Indexed: 01/09/2023]
Abstract
Respiratory diseases are a major reason for death in both men and women worldwide. The development of therapies for these diseases has been slow and the lack of relevant human models to understand lung biology inhibits therapeutic discovery. The lungs are structurally and functionally complex with many different cell types which makes designing relevant lung models particularly challenging. The traditional two-dimensional (2D) cell line cultures are, therefore, not a very accurate representation of the in vivo lung tissue. The recent development of three-dimensional (3D) co-culture systems, popularly known as organoids/spheroids, aims to bridge the gap between 'in-dish' and 'in-tissue' cell behavior. These 3D cultures are modeling systems that are widely divergent in terms of culturing techniques (bottom-up/top-down) that can be developed from stem cells (adult/embryonic/pluripotent stem cells), primary cells or from two or more types of cells, to build a co-culture system. Lung 3D models have diverse applications including the understanding of lung development, lung regeneration, disease modeling, compound screening, and personalized medicine. In this review, we discuss the different techniques currently being used to generate 3D models and their associated cellular and biological materials. We further detail the potential applications of lung 3D cultures for disease modeling and advances in throughput for drug screening.
Collapse
|
27
|
Mir M, Chen J, Pinezich MR, O'Neill JD, Huang SXL, Vunjak-Novakovic G, Kim J. Imaging-guided bioreactor for de-epithelialization and long-term cultivation of ex vivo rat trachea. LAB ON A CHIP 2022; 22:1018-1031. [PMID: 35166739 PMCID: PMC8942046 DOI: 10.1039/d1lc01105g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Recent synergistic advances in organ-on-chip and tissue engineering technologies offer opportunities to create in vitro-grown tissue or organ constructs that can faithfully recapitulate their in vivo counterparts. Such in vitro tissue or organ constructs can be utilized in multiple applications, including rapid drug screening, high-fidelity disease modeling, and precision medicine. Here, we report an imaging-guided bioreactor that allows in situ monitoring of the lumen of ex vivo airway tissues during controlled in vitro tissue manipulation and cultivation of isolated rat trachea. Using this platform, we demonstrated partial removal of the rat tracheal epithelium (i.e., de-epithelialization) without disrupting the underlying subepithelial cells and extracellular matrix. Through different tissue evaluation assays, such as immunofluorescent staining, DNA/protein quantification, and electron beam microscopy, we showed that the epithelium of the tracheal lumen can be effectively removed with negligible disruption in the underlying tissue layers, such as cartilage and blood vessel. Notably, using a custom-built micro-optical imaging device integrated with the bioreactor, the trachea lumen was visualized at the cellular level, and removal of the endogenous epithelium and distribution of locally delivered exogenous cells were demonstrated in situ. Moreover, the de-epithelialized trachea supported on the bioreactor allowed attachment and growth of exogenous cells seeded topically on its denuded tissue surface. Collectively, the results suggest that our imaging-enabled rat trachea bioreactor and localized cell replacement method can facilitate creation of bioengineered in vitro airway tissue that can be used in different biomedical applications.
Collapse
Affiliation(s)
- Mohammad Mir
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA.
| | - Jiawen Chen
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA.
| | - Meghan R Pinezich
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - John D O'Neill
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Sarah X L Huang
- Center for Stem Cell and Regenerative Medicine, University of Texas Health Science Center, Houston, TX, USA
| | | | - Jinho Kim
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA.
| |
Collapse
|
28
|
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: 2.8] [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
|
29
|
Vindin HJ, Oliver BG, Weiss AS. Elastin in healthy and diseased lung. Curr Opin Biotechnol 2021; 74:15-20. [PMID: 34781101 DOI: 10.1016/j.copbio.2021.10.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/12/2021] [Accepted: 10/19/2021] [Indexed: 01/05/2023]
Abstract
Elastic fibers are an essential part of the pulmonary extracellular matrix (ECM). Intact elastin is required for normal function and its damage contributes profoundly to the etiology and pathology of lung disease. This highlights the need for novel lung-specific imaging methodology that enables high-resolution 3D visualization of the ECM. We consider elastin's involvement in chronic respiratory disease and examine recent methods for imaging and modeling of the lung in the context of advances in lung tissue engineering for research and clinical application.
Collapse
Affiliation(s)
- Howard J Vindin
- Charles Perkins Centre, The University of Sydney, Sydney 2006, NSW, Australia; School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia; The Woolcock Institute, The University of Sydney, Sydney 2006, NSW, Australia
| | - Brian Gg Oliver
- The Woolcock Institute, The University of Sydney, Sydney 2006, NSW, Australia
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, Sydney 2006, NSW, Australia; School of Life and Environmental Sciences, The University of Sydney, 2006 Sydney, NSW, Australia; Sydney Nano Institute, The University of Sydney, 2006 Sydney, NSW, Australia.
| |
Collapse
|
30
|
Singh AV, Romeo A, Scott K, Wagener S, Leibrock L, Laux P, Luch A, Kerkar P, Balakrishnan S, Dakua SP, Park B. Emerging Technologies for In Vitro Inhalation Toxicology. Adv Healthc Mater 2021; 10:e2100633. [PMID: 34292676 PMCID: PMC11468957 DOI: 10.1002/adhm.202100633] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/04/2021] [Indexed: 12/20/2022]
Abstract
Respiratory toxicology remains a major research area in the 21st century since current scenario of airborne viral infection transmission and pollutant inhalation is expected to raise the annual morbidity beyond 2 million. Clinical and epidemiological research connecting human exposure to air contaminants to understand adverse pulmonary health outcomes is, therefore, an immediate subject of human health assessment. Important observations in defining systemic effects of environmental contaminants on inhalation metabolic dysfunction, liver health, and gastrointestinal tract have been well explored with in vivo models. In this review, a framework is provided, a paradigm is established about inhalation toxicity testing in vitro, and a brief overview of breathing Lungs-on-Chip (LoC) as design concepts is given. The optimized bioengineering approaches and microfluidics with their fundamental pros, and cons are presented. There are different strategies that researchers apply to inhalation toxicity studies to assess a variety of inhalable substances and relevant LoC approaches. A case study from published literature and frame arguments about reproducibility as well as in vitro/in vivo correlations are discussed. Finally, the opportunities and challenges in soft robotics, systems inhalation toxicology approach integrating bioengineering, machine learning, and artificial intelligence to address a multitude model for future toxicology are discussed.
Collapse
Affiliation(s)
- Ajay Vikram Singh
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)Max‐Dohrn‐Strasse 8‐10Berlin10589Germany
| | - Anthony Romeo
- Department of Chemical EngineeringRayen School of EngineeringYoungstown State UniversityYoungstownOH44555USA
| | - Kassandra Scott
- Department of Chemical EngineeringRayen School of EngineeringYoungstown State UniversityYoungstownOH44555USA
| | - Sandra Wagener
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)Max‐Dohrn‐Strasse 8‐10Berlin10589Germany
| | - Lars Leibrock
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)Max‐Dohrn‐Strasse 8‐10Berlin10589Germany
| | - Peter Laux
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)Max‐Dohrn‐Strasse 8‐10Berlin10589Germany
| | - Andreas Luch
- Department of Chemical and Product SafetyGerman Federal Institute for Risk Assessment (BfR)Max‐Dohrn‐Strasse 8‐10Berlin10589Germany
| | - Pranali Kerkar
- ICMR – National AIDS Research Institute (NARI)PuneMaharashtra411026India
| | | | | | - Byung‐Wook Park
- Department of Chemical EngineeringRayen School of EngineeringYoungstown State UniversityYoungstownOH44555USA
| |
Collapse
|
31
|
Liu Y, Sun L, Zhang H, Shang L, Zhao Y. Microfluidics for Drug Development: From Synthesis to Evaluation. Chem Rev 2021; 121:7468-7529. [PMID: 34024093 DOI: 10.1021/acs.chemrev.0c01289] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Drug development is a long process whose main content includes drug synthesis, drug delivery, and drug evaluation. Compared with conventional drug development procedures, microfluidics has emerged as a revolutionary technology in that it offers a miniaturized and highly controllable environment for bio(chemical) reactions to take place. It is also compatible with analytical strategies to implement integrated and high-throughput screening and evaluations. In this review, we provide a comprehensive summary of the entire microfluidics-based drug development system, from drug synthesis to drug evaluation. The challenges in the current status and the prospects for future development are also discussed. We believe that this review will promote communications throughout diversified scientific and engineering communities that will continue contributing to this burgeoning field.
Collapse
Affiliation(s)
- Yuxiao Liu
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyu Sun
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Hui Zhang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Luoran Shang
- Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
32
|
Ahmed HMMAM, Moreira Teixeira LS. New Endeavors of (Micro)Tissue Engineering: Cells Tissues Organs on-Chip and Communication Thereof. Cells Tissues Organs 2021; 211:721-735. [PMID: 34198305 DOI: 10.1159/000516356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 03/30/2021] [Indexed: 01/25/2023] Open
Abstract
The development of new therapies is tremendously hampered by the insufficient availability of human model systems suitable for preclinical research on disease target identification, drug efficacy, and toxicity. Thus, drug failures in clinical trials are too common and too costly. Animal models or standard 2D in vitro tissue cultures, regardless of whether they are human based, are regularly not representative of specific human responses. Approaching near human tissues and organs test systems is the key goal of organs-on-chips (OoC) technology. This technology is currently showing its potential to reduce both drug development costs and time-to-market, while critically lessening animal testing. OoC are based on human (stem) cells, potentially derived from healthy or disease-affected patients, thereby amenable to personalized therapy development. It is noteworthy that the OoC market potential goes beyond pharma, with the possibility to test cosmetics, food additives, or environmental contaminants. This (micro)tissue engineering-based technology is highly multidisciplinary, combining fields such as (developmental) biology, (bio)materials, microfluidics, sensors, and imaging. The enormous potential of OoC is currently facing an exciting new challenge: emulating cross-communication between tissues and organs, to simulate more complex systemic responses, such as in cancer, or restricted to confined environments, as occurs in osteoarthritis. This review describes key examples of multiorgan/tissue-on-chip approaches, or linked organs/tissues-on-chip, focusing on challenges and promising new avenues of this advanced model system. Additionally, major emphasis is given to the translation of established tissue engineering approaches, bottom up and top down, towards the development of more complex, robust, and representative (multi)organ/tissue-on-chip approaches.
Collapse
Affiliation(s)
- Haysam M M A M Ahmed
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands,
| | - Liliana S Moreira Teixeira
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, The Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
33
|
Bennet TJ, Randhawa A, Hua J, Cheung KC. Airway-On-A-Chip: Designs and Applications for Lung Repair and Disease. Cells 2021; 10:1602. [PMID: 34206722 PMCID: PMC8304815 DOI: 10.3390/cells10071602] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022] Open
Abstract
The lungs are affected by illnesses including asthma, chronic obstructive pulmonary disease, and infections such as influenza and SARS-CoV-2. Physiologically relevant models for respiratory conditions will be essential for new drug development. The composition and structure of the lung extracellular matrix (ECM) plays a major role in the function of the lung tissue and cells. Lung-on-chip models have been developed to address some of the limitations of current two-dimensional in vitro models. In this review, we describe various ECM substitutes utilized for modeling the respiratory system. We explore the application of lung-on-chip models to the study of cigarette smoke and electronic cigarette vapor. We discuss the challenges and opportunities related to model characterization with an emphasis on in situ characterization methods, both established and emerging. We discuss how further advancements in the field, through the incorporation of interstitial cells and ECM, have the potential to provide an effective tool for interrogating lung biology and disease, especially the mechanisms that involve the interstitial elements.
Collapse
Affiliation(s)
- Tanya J. Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Avineet Randhawa
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Hua
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
34
|
Khedoe PPPSJ, Wu X, Gosens R, Hiemstra PS. Repairing damaged lungs using regenerative therapy. Curr Opin Pharmacol 2021; 59:85-94. [PMID: 34161852 PMCID: PMC9188766 DOI: 10.1016/j.coph.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/14/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022]
Abstract
There is an urgent need for better treatment of lung diseases that are a major cause of morbidity and mortality worldwide. This urgency is illustrated by the current COVID-19 health crisis. Moderate-to-extensive lung injury characterizes several lung diseases, and not only therapies that reduce such lung injury are needed but also those that regenerate lung tissue and repair existing lung injury. At present, such therapies are not available, but as a result of a rapid increase in our understanding of lung development and repair, lung regenerative therapies are on the horizon. Here, we discuss existing targets for treatment, as well as novel strategies for development of pharmacological and cell therapy-based regenerative treatment for a variety of lung diseases and clinical studies. We discuss how both patient-relevant in vitro disease models using innovative culture techniques and other advanced new technologies aid in the development of pulmonary regenerative medicine.
Collapse
Affiliation(s)
| | - Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
35
|
Pellegrino E, Gutierrez MG. Human stem cell-based models for studying host-pathogen interactions. Cell Microbiol 2021; 23:e13335. [PMID: 33792137 DOI: 10.1111/cmi.13335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
The use of human cell lines and primary cells as in vitro models represents a valuable approach to study cellular responses to infection. However, with the advent of new molecular technologies and tools available, there is a growing need to develop more physiologically relevant systems to overcome cell line model limitations and better mimic human disease. Since the discovery of human stem cells, its use has revolutionised the development of in vitro models. This is because after differentiation, these cells have the potential to reflect in vivo cell phenotypes and allow for probing questions in numerous fields of the biological sciences. Moreover, the possibility to combine the advantages of stem cell-derived cell types with genome editing technologies and engineered 3D microenvironments, provides enormous potential for producing in vitro systems to investigate cellular responses to infection that are both relevant and predictive. Here, we discuss recent advances in the use of human stem cells to model host-pathogen interactions, highlighting emerging technologies in the field of stem cell biology that can be exploited to investigate the fundamental biology of infection. TAKE AWAYS: hPSC overcome current limitations to study host-pathogen interactions in vitro. Genome editing can be used in hPSC to study cellular responses to infection. hPSC, 3D models and genome editing can recreate physiological in vitro systems.
Collapse
Affiliation(s)
- Enrica Pellegrino
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
36
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
37
|
Aich A, Lamarre Y, Sacomani DP, Kashima S, Covas DT, de la Torre LG. Microfluidics in Sickle Cell Disease Research: State of the Art and a Perspective Beyond the Flow Problem. Front Mol Biosci 2021; 7:558982. [PMID: 33763448 PMCID: PMC7982466 DOI: 10.3389/fmolb.2020.558982] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/24/2020] [Indexed: 01/21/2023] Open
Abstract
Sickle cell disease (SCD) is the monogenic hemoglobinopathy where mutated sickle hemoglobin molecules polymerize to form long fibers under deoxygenated state and deform red blood cells (RBCs) into predominantly sickle form. Sickled RBCs stick to the vascular bed and obstruct blood flow in extreme conditions, leading to acute painful vaso-occlusion crises (VOCs) – the leading cause of mortality in SCD. Being a blood disorder of deformed RBCs, SCD manifests a wide-range of organ-specific clinical complications of life (in addition to chronic pain) such as stroke, acute chest syndrome (ACS) and pulmonary hypertension in the lung, nephropathy, auto-splenectomy, and splenomegaly, hand-foot syndrome, leg ulcer, stress erythropoiesis, osteonecrosis and osteoporosis. The physiological inception for VOC was initially thought to be only a fluid flow problem in microvascular space originated from increased viscosity due to aggregates of sickled RBCs; however, over the last three decades, multiple molecular and cellular mechanisms have been identified that aid the VOC in vivo. Activation of adhesion molecules in vascular endothelium and on RBC membranes, activated neutrophils and platelets, increased viscosity of the blood, and fluid physics driving sickled and deformed RBCs to the vascular wall (known as margination of flow) – all of these come together to orchestrate VOC. Microfluidic technology in sickle research was primarily adopted to benefit from mimicking the microvascular network to observe RBC flow under low oxygen conditions as models of VOC. However, over the last decade, microfluidics has evolved as a valuable tool to extract biophysical characteristics of sickle red cells, measure deformability of sickle red cells under simulated oxygen gradient and shear, drug testing, in vitro models of intercellular interaction on endothelialized or adhesion molecule-functionalized channels to understand adhesion in sickle microenvironment, characterizing biomechanics and microrheology, biomarker identification, and last but not least, for developing point-of-care diagnostic technologies for low resource setting. Several of these platforms have already demonstrated true potential to be translated from bench to bedside. Emerging microfluidics-based technologies for studying heterotypic cell–cell interactions, organ-on-chip application and drug dosage screening can be employed to sickle research field due to their wide-ranging advantages.
Collapse
Affiliation(s)
- Anupam Aich
- Intel Corporation, Hillsboro, OR, United States
| | - Yann Lamarre
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniel Pereira Sacomani
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, Brazil
| | - Simone Kashima
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
38
|
Zamprogno P, Thoma G, Cencen V, Ferrari D, Putz B, Michler J, Fantner GE, Guenat OT. Mechanical Properties of Soft Biological Membranes for Organ-on-a-Chip Assessed by Bulge Test and AFM. ACS Biomater Sci Eng 2021; 7:2990-2997. [PMID: 33651947 DOI: 10.1021/acsbiomaterials.0c00515] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Advanced in vitro models called "organ-on-a-chip" can mimic the specific cellular environment found in various tissues. Many of these models include a thin, sometimes flexible, membrane aimed at mimicking the extracellular matrix (ECM) scaffold of in vivo barriers. These membranes are often made of polydimethylsiloxane (PDMS), a silicone rubber that poorly mimics the chemical and physical properties of the basal membrane. However, the ECM and its mechanical properties play a key role in the homeostasis of a tissue. Here, we report about biological membranes with a composition and mechanical properties similar to those found in vivo. Two types of collagen-elastin (CE) membranes were produced: vitrified and nonvitrified (called "hydrogel membrane"). Their mechanical properties were characterized using the bulge test method. The results were compared using atomic force microscopy (AFM), a standard technique used to evaluate the Young's modulus of soft materials at the nanoscale. Our results show that CE membranes with stiffnesses ranging from several hundred of kPa down to 1 kPa can be produced by tuning the CE ratio, the production mode (vitrified or not), and/or certain parameters such as temperature. The Young's modulus can easily be determined using the bulge test. This method is a robust and reproducible to determine membrane stiffness, even for soft membranes, which are more difficult to assess by AFM. Assessment of the impact of substrate stiffness on the spread of human fibroblasts on these surfaces showed that cell spread is lower on softer surfaces than on stiffer surfaces.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Veronika Cencen
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Barbara Putz
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Johann Michler
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Georg E Fantner
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern 3008, Switzerland.,Department of General Thoracic Surgery, University Hospital of Bern, Bern 3008, Switzerland
| |
Collapse
|
39
|
Evans JF, Obraztsova K, Lin SM, Krymskaya VP. CrossTORC and WNTegration in Disease: Focus on Lymphangioleiomyomatosis. Int J Mol Sci 2021; 22:ijms22052233. [PMID: 33668092 PMCID: PMC7956553 DOI: 10.3390/ijms22052233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) and wingless-related integration site (Wnt) signal transduction networks are evolutionarily conserved mammalian growth and cellular development networks. Most cells express many of the proteins in both pathways, and this review will briefly describe only the key proteins and their intra- and extracellular crosstalk. These complex interactions will be discussed in relation to cancer development, drug resistance, and stem cell exhaustion. This review will also highlight the tumor-suppressive tuberous sclerosis complex (TSC) mutated, mTOR-hyperactive lung disease of women, lymphangioleiomyomatosis (LAM). We will summarize recent advances in the targeting of these pathways by monotherapy or combination therapy, as well as future potential treatments.
Collapse
|
40
|
Duzagac F, Saorin G, Memeo L, Canzonieri V, Rizzolio F. Microfluidic Organoids-on-a-Chip: Quantum Leap in Cancer Research. Cancers (Basel) 2021; 13:737. [PMID: 33578886 PMCID: PMC7916612 DOI: 10.3390/cancers13040737] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Organ-like cell clusters, so-called organoids, which exhibit self-organized and similar organ functionality as the tissue of origin, have provided a whole new level of bioinspiration for ex vivo systems. Microfluidic organoid or organs-on-a-chip platforms are a new group of micro-engineered promising models that recapitulate 3D tissue structure and physiology and combines several advantages of current in vivo and in vitro models. Microfluidics technology is used in numerous applications since it allows us to control and manipulate fluid flows with a high degree of accuracy. This system is an emerging tool for understanding disease development and progression, especially for personalized therapeutic strategies for cancer treatment, which provide well-grounded, cost-effective, powerful, fast, and reproducible results. In this review, we highlight how the organoid-on-a-chip models have improved the potential of efficiency and reproducibility of organoid cultures. More widely, we discuss current challenges and development on organoid culture systems together with microfluidic approaches and their limitations. Finally, we describe the recent progress and potential utilization in the organs-on-a-chip practice.
Collapse
Affiliation(s)
- Fahriye Duzagac
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy;
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
41
|
Zamprogno P, Wüthrich S, Achenbach S, Thoma G, Stucki JD, Hobi N, Schneider-Daum N, Lehr CM, Huwer H, Geiser T, Schmid RA, Guenat OT. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol 2021; 4:168. [PMID: 33547387 PMCID: PMC7864995 DOI: 10.1038/s42003-021-01695-0] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Simon Wüthrich
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Sven Achenbach
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Janick D Stucki
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nina Hobi
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nicole Schneider-Daum
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Hanno Huwer
- SHG Clinics, Department of Cardiothoracic Surgery, Völklingen Heart Center, Völklingen, Germany
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland.
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
42
|
Khedoe P, Marges E, Hiemstra P, Ninaber M, Geelhoed M. Interstitial Lung Disease in Patients With Systemic Sclerosis: Toward Personalized-Medicine-Based Prediction and Drug Screening Models of Systemic Sclerosis-Related Interstitial Lung Disease (SSc-ILD). Front Immunol 2020; 11:1990. [PMID: 33013852 PMCID: PMC7500178 DOI: 10.3389/fimmu.2020.01990] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 07/23/2020] [Indexed: 12/16/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disease, characterized by immune dysregulation and progressive fibrosis. Interstitial lung disease (ILD) is the most common cause of death among SSc patients and there are currently very limited approved disease-modifying treatment options for systemic sclerosis-related interstitial lung disease (SSc-ILD). The mechanisms underlying pulmonary fibrosis in SSc-ILD are not completely unraveled, and knowledge on fibrotic processes has been acquired mostly from studies in idiopathic pulmonary fibrosis (IPF). The incomplete knowledge of SSc-ILD pathogenesis partly explains the limited options for disease-modifying therapy for SSc-ILD. Fibrosis in IPF appears to be related to aberrant repair following injury, but whether this also holds for SSc-ILD is less evident. Furthermore, immune dysregulation appears to contribute to pro-fibrotic responses in SSc-ILD, perhaps more than in IPF. In addition, SSc-ILD patient heterogeneity complicates the understanding of the underlying mechanisms of disease development, and more importantly, limits correct clinical diagnosis and treatment effectivity. Therefore, there is an unmet need for patient-relevant (in vitro) models to examine patient-specific disease pathogenesis, predict disease progression, screen appropriate treatment regimens and identify new targets for treatment. Technological advances in in vitro patient-relevant disease modeling, including (human induced pluripotent stem cell (hiPSC)-derived) lung epithelial cells, organoids and organ-on-chip technology offer a platform that has the potential to contribute to unravel the underlying mechanisms of SSc-ILD development. Combining these models with state-of-the-art analysis platforms, including (single cell) RNA sequencing and (imaging) mass cytometry, may help to delineate pathogenic mechanisms and define new treatment targets of SSc-ILD.
Collapse
Affiliation(s)
- Padmini Khedoe
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Emiel Marges
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Pieter Hiemstra
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Maarten Ninaber
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Miranda Geelhoed
- Department of Pulmonology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| |
Collapse
|
43
|
Host-Pathogen Responses to Pandemic Influenza H1N1pdm09 in a Human Respiratory Airway Model. Viruses 2020; 12:v12060679. [PMID: 32599823 PMCID: PMC7354428 DOI: 10.3390/v12060679] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023] Open
Abstract
The respiratory Influenza A Viruses (IAVs) and emerging zoonotic viruses such as Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) pose a significant threat to human health. To accelerate our understanding of the host–pathogen response to respiratory viruses, the use of more complex in vitro systems such as normal human bronchial epithelial (NHBE) cell culture models has gained prominence as an alternative to animal models. NHBE cells were differentiated under air-liquid interface (ALI) conditions to form an in vitro pseudostratified epithelium. The responses of well-differentiated (wd) NHBE cells were examined following infection with the 2009 pandemic Influenza A/H1N1pdm09 strain or following challenge with the dsRNA mimic, poly(I:C). At 30 h postinfection with H1N1pdm09, the integrity of the airway epithelium was severely impaired and apical junction complex damage was exhibited by the disassembly of zona occludens-1 (ZO-1) from the cell cytoskeleton. wdNHBE cells produced an innate immune response to IAV-infection with increased transcription of pro- and anti-inflammatory cytokines and chemokines and the antiviral viperin but reduced expression of the mucin-encoding MUC5B, which may impair mucociliary clearance. Poly(I:C) produced similar responses to IAV, with the exception of MUC5B expression which was more than 3-fold higher than for control cells. This study demonstrates that wdNHBE cells are an appropriate ex-vivo model system to investigate the pathogenesis of respiratory viruses.
Collapse
|
44
|
Yang JW, Shen YC, Lin KC, Cheng SJ, Chen SL, Chen CY, Kumar PV, Lin SF, Lu HE, Chen GY. Organ-on-a-Chip: Opportunities for Assessing the Toxicity of Particulate Matter. Front Bioeng Biotechnol 2020; 8:519. [PMID: 32548105 PMCID: PMC7272695 DOI: 10.3389/fbioe.2020.00519] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/01/2020] [Indexed: 12/25/2022] Open
Abstract
Recent developments in epidemiology have confirmed that airborne particulates are directly associated with respiratory pathology and mortality. Although clinical studies have yielded evidence of the effects of many types of fine particulates on human health, it still does not have a complete understanding of how physiological reactions are caused nor to the changes and damages associated with cellular and molecular mechanisms. Currently, most health assessment studies of particulate matter (PM) are conducted through cell culture or animal experiments. The results of such experiments often do not correlate with clinical findings or actual human reactions, and they also cause difficulty when investigating the causes of air pollution and associated human health hazards, the analysis of biomarkers, and the development of future pollution control strategies. Microfluidic-based cell culture technology has considerable potential to expand the capabilities of conventional cell culture by providing high-precision measurement, considerably increasing the potential for the parallelization of cellular assays, ensuring inexpensive automation, and improving the response of the overall cell culture in a more physiologically relevant context. This review paper focuses on integrating the important respiratory health problems caused by air pollution today, as well as the development and application of biomimetic organ-on-a-chip technology. This more precise experimental model is expected to accelerate studies elucidating the effect of PM on the human body and to reveal new opportunities for breakthroughs in disease research and drug development.
Collapse
Affiliation(s)
- Jia-Wei Yang
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Chih Shen
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan.,Ph.D. Degree Program of Biomedical Science and Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Ko-Chih Lin
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Sheng-Jen Cheng
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Shiue-Luen Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Chong-You Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Priyank V Kumar
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Shien-Fong Lin
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Huai-En Lu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering National Chiao Tung University, Hsinchu, Taiwan.,Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
45
|
Broadbent L, Manzoor S, Zarcone MC, Barabas J, Shields MD, Saglani S, Lloyd CM, Bush A, Custovic A, Ghazal P, Gore M, Marsland B, Roberts G, Schwarze J, Turner S, Power UF. Comparative primary paediatric nasal epithelial cell culture differentiation and RSV-induced cytopathogenesis following culture in two commercial media. PLoS One 2020; 15:e0228229. [PMID: 32214336 PMCID: PMC7098550 DOI: 10.1371/journal.pone.0228229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/05/2020] [Indexed: 02/02/2023] Open
Abstract
The culture of differentiated human airway epithelial cells allows the study of pathogen-host interactions and innate immune responses in a physiologically relevant in vitro model. As the use of primary cell culture has gained popularity the availability of the reagents needed to generate these cultures has increased. In this study we assessed two different media, Promocell and PneumaCult, during the differentiation and maintenance of well-differentiated primary nasal epithelial cell cultures (WD-PNECs). We compared and contrasted the consequences of these media on WD-PNEC morphological and physiological characteristics and their responses to respiratory syncytial virus (RSV) infection. We found that cultures generated using PneumaCult resulted in greater total numbers of smaller, tightly packed, pseudostratified cells. However, cultures from both media resulted in similar proportions of ciliated and goblet cells. There were no differences in RSV growth kinetics, although more ciliated cells were infected in the PneumaCult cultures. There was also significantly more IL-29/IFNλ1 secreted from PneumaCult compared to Promocell cultures following infection. In conclusion, the type of medium used for the differentiation of primary human airway epithelial cells may impact experimental results.
Collapse
Affiliation(s)
- Lindsay Broadbent
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Sheerien Manzoor
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Maria C. Zarcone
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, England, United Kingdom
| | - Judit Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Michael D. Shields
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, Northern Ireland, United Kingdom
- Royal Belfast Hospital for Sick Children, Belfast Health & Social Care Trust, Belfast, Northern Ireland, United Kingdom
| | - Sejal Saglani
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, England, United Kingdom
| | - Claire M. Lloyd
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, England, United Kingdom
| | - Andrew Bush
- Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, England, United Kingdom
| | - Adnan Custovic
- Department of Paediatrics, Imperial College London, London, England, United Kingdom
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, Deanery of Biomedical Sciences, University of Edinburgh Medical School, Edinburgh, Scotland, United Kingdom
| | - Mindy Gore
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College, London, England, United Kingdom
| | - Ben Marsland
- Department of Immunology and Pathology, Monash University, Melbourne, Scotland, Australia
| | - Graham Roberts
- Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, England, United Kingdom
| | - Jurgen Schwarze
- Child Life and Health and MRC-Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Steve Turner
- Child Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Ultan F. Power
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
46
|
Artzy-Schnirman A, Lehr CM, Sznitman J. Advancing human in vitro pulmonary disease models in preclinical research: opportunities for lung-on-chips. Expert Opin Drug Deliv 2020; 17:621-625. [DOI: 10.1080/17425247.2020.1738380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa, Israel
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarland University, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion – Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Shrestha J, Razavi Bazaz S, Aboulkheyr Es H, Yaghobian Azari D, Thierry B, Ebrahimi Warkiani M, Ghadiri M. Lung-on-a-chip: the future of respiratory disease models and pharmacological studies. Crit Rev Biotechnol 2020; 40:213-230. [PMID: 31906727 DOI: 10.1080/07388551.2019.1710458] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recently, organ-on-a-chip models, which are microfluidic devices that mimic the cellular architecture and physiological environment of an organ, have been developed and extensively investigated. The chips can be tailored to accommodate the disease conditions pertaining to many organs; and in the case of this review, the lung. Lung-on-a-chip models result in a more accurate reflection compared to conventional in vitro models. Pharmaceutical drug testing methods traditionally use animal models in order to evaluate pharmacological and toxicological responses to a new agent. However, these responses do not directly reflect human physiological responses. In this review, current and future applications of the lung-on-a-chip in the respiratory system will be discussed. Furthermore, the limitations of current conventional in vitro models used for respiratory disease modeling and drug development will be addressed. Highlights of additional translational aspects of the lung-on-a-chip will be discussed in order to demonstrate the importance of this subject for medical research.
Collapse
Affiliation(s)
- Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia.,Faculty of Medicine and Health, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | | | | | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence in Convergent Bio and Nano Science and Technology, University of South Australia, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia.,Institute of Molecular Medicine, Sechenov University, Moscow, Russia
| | - Maliheh Ghadiri
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia.,Faculty of Medicine and Health, Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia.,School of Medicine and Public Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
48
|
Abstract
As the prevalence and impact of lung diseases continue to increase worldwide, new therapeutic strategies are desperately needed. Advances in lung-regenerative medicine, a broad field encompassing stem cells, cell-based therapies, and a range of bioengineering approaches, offer new insights into and new techniques for studying lung physiology and pathophysiology. This provides a platform for the development of novel therapeutic approaches. Applicability to chronic obstructive pulmonary disease of recent advances and applications in cell-based therapies, predominantly those with mesenchymal stromal cell-based approaches, and bioengineering approaches for lung diseases are reviewed.
Collapse
|
49
|
Hiemstra PS, Tetley TD, Janes SM. Airway and alveolar epithelial cells in culture. Eur Respir J 2019; 54:13993003.00742-2019. [DOI: 10.1183/13993003.00742-2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/13/2019] [Indexed: 12/29/2022]
|
50
|
Glaser L, Coulter PJ, Shields M, Touzelet O, Power UF, Broadbent L. Airway Epithelial Derived Cytokines and Chemokines and Their Role in the Immune Response to Respiratory Syncytial Virus Infection. Pathogens 2019; 8:E106. [PMID: 31331089 PMCID: PMC6789711 DOI: 10.3390/pathogens8030106] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022] Open
Abstract
The airway epithelium is the primary target of respiratory syncytial virus infection. It is an important component of the antiviral immune response. It contributes to the recruitment and activation of innate immune cells from the periphery through the secretion of cytokines and chemokines. This paper provides a broad review of the cytokines and chemokines secreted from human airway epithelial cell models during respiratory syncytial virus (RSV) infection based on a comprehensive literature review. Epithelium-derived chemokines constitute most inflammatory mediators secreted from the epithelium during RSV infection. This suggests chemo-attraction of peripheral immune cells, such as monocytes, neutrophils, eosinophils, and natural killer cells as a key function of the epithelium. The reports of epithelium-derived cytokines are limited. Recent research has started to identify novel cytokines, the functions of which remain largely unknown in the wider context of the RSV immune response. It is argued that the correct choice of in vitro models used for investigations of epithelial immune functions during RSV infection could facilitate greater progress in this field.
Collapse
Affiliation(s)
- Lena Glaser
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK
| | - Patricia J Coulter
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK
- Department of Paediatric Respiratory Medicine, Royal Belfast Hospital for Sick Children, Belfast BT12 6BE, Northern Ireland, UK
| | - Michael Shields
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK
- Department of Paediatric Respiratory Medicine, Royal Belfast Hospital for Sick Children, Belfast BT12 6BE, Northern Ireland, UK
| | - Olivier Touzelet
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK
| | - Ultan F Power
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK.
| | - Lindsay Broadbent
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast BT9 7BL, Northern Ireland, UK.
| |
Collapse
|