1
|
Mendes M, Morais AS, Carlos A, Sousa JJ, Pais AC, Mihăilă SM, Vitorino C. Organ-on-a-chip: Quo vademus? Applications and regulatory status. Colloids Surf B Biointerfaces 2025; 249:114507. [PMID: 39826309 DOI: 10.1016/j.colsurfb.2025.114507] [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: 09/29/2024] [Revised: 12/15/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Organ-on-a-chip systems, also referred to as microphysiological systems (MPS), represent an advance in bioengineering microsystems designed to mimic key aspects of human organ physiology and function. Drawing inspiration from the intricate and hierarchical architecture of the human body, these innovative platforms have emerged as invaluable in vitro tools with wide-ranging applications in drug discovery and development, as well as in enhancing our understanding of disease physiology. The facility to replicate human tissues within physiologically relevant three-dimensional multicellular environments empowers organ-on-a-chip systems with versatility throughout different stages of the drug development process. Moreover, these systems can be tailored to mimic specific disease states, facilitating the investigation of disease progression, drug responses, and potential therapeutic interventions. In particular, they can demonstrate, in early-phase pre-clinical studies, the safety and toxicity profiles of potential therapeutic compounds. Furthermore, they play a pivotal role in the in vitro evaluation of drug efficacy and the modeling of human diseases. One of the most promising prospects of organ-on-a-chip technology is to simulate the pathophysiology of specific subpopulations and even individual patients, thereby being used in personalized medicine. By mimicking the physiological responses of diverse patient groups, these systems hold the promise of revolutionizing therapeutic strategies, guiding them towards tailored intervention to the unique needs of each patient. This review presents the development status and evolution of microfluidic platforms that have facilitated the transition from cells to organs recreated on chips and some of the opportunities and applications offered by organ-on-a-chip technology. Additionally, the current potential and future perspectives of these microphysiological systems and the challenges this technology still faces are discussed.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Ana Sofia Morais
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Carlos
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - João José Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Alberto Canelas Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal
| | - Silvia M Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3000-535, Portugal.
| |
Collapse
|
2
|
Bhaskar V, Kumar R, Praharaj MR, Gandham S, Maity HK, Sarkar U, Dey B. A bovine pulmosphere model and multiomics reveal early host response signature in tuberculosis. Commun Biol 2025; 8:559. [PMID: 40186000 PMCID: PMC11971429 DOI: 10.1038/s42003-025-07883-6] [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: 04/17/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025] Open
Abstract
Early interactions between tubercle bacilli and lung cells are critical in tuberculosis (TB) pathogenesis. Conventional two-dimensional cell cultures fail to replicate the multicellular complexity of lungs. We introduce a three-dimensional pulmosphere model for Mycobacterium tuberculosis infection in bovine systems, demonstrating through comprehensive transcriptome and proteome analyses that these multicellular spheroids closely mimic lung cell diversity, interactions, and extracellular matrix (ECM) composition. Cell viability, hypoxia, and reactive oxygen species assessments over three weeks confirm the model's suitability. To establish infection, we employed M. bovis BCG-an attenuated vaccine strain, and M. tuberculosis H37Rv-a laboratory adapted human clinical strain that is attenuated for cattle infection compared to M. bovis. Both infection upregulated key host pathways; however, M. tuberculosis induced distinct responses, including enhanced ECM receptors expression, neutrophil chemotaxis, interferon signaling, and RIG-1 signaling. A six genes/protein signature- IRF1, CCL5, CXCL8, CXCL10, SERPINE1, and CFB -emerges as an early host response marker to M. tuberculosis infection. Infection with virulent M. bovis and M. orygis revealed a shared upregulated gene signature across Mycobacterium tuberculosis complex species, but with pathogen-specific variations. This study presents a robust ex vivo bovine pulmosphere TB model with implications in biomarkers discovery, high-throughput drug screening, and TB control strategies.
Collapse
Affiliation(s)
- Vinay Bhaskar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
| | - Rishi Kumar
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Manas Ranjan Praharaj
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Sripratyusha Gandham
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Hemanta Kumar Maity
- Department of Avian Sciences, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Uttam Sarkar
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Bappaditya Dey
- National Institute of Animal Biotechnology, Hyderabad, Telangana, India.
- Regional Centre for Biotechnology, Faridabad, Haryana, India.
| |
Collapse
|
3
|
Kalulu M, Chilikwazi B, Hu J, Fu G. Soft Actuators and Actuation: Design, Synthesis, and Applications. Macromol Rapid Commun 2025; 46:e2400282. [PMID: 38850266 DOI: 10.1002/marc.202400282] [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: 04/29/2024] [Revised: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Soft actuators are one of the most promising technological advancements with potential solutions to diverse fields' day-to-day challenges. Soft actuators derived from hydrogel materials possess unique features such as flexibility, responsiveness to stimuli, and intricate deformations, making them ideal for soft robotics, artificial muscles, and biomedical applications. This review provides an overview of material composition and design techniques for hydrogel actuators, exploring 3D printing, photopolymerization, cross-linking, and microfabrication methods for improved actuation. It examines applications of hydrogel actuators in biomedical, soft robotics, bioinspired systems, microfluidics, lab-on-a-chip devices, and environmental, and energy systems. Finally, it discusses challenges, opportunities, advancements, and regulatory aspects related to hydrogel actuators.
Collapse
Affiliation(s)
- Mulenga Kalulu
- School of Chemistry and Chemical Engineering, Southeast University, Jiangning, Nanjing, Jiangsu Province, 211189, P. R. China
- Department of Chemistry, School of Natural Sciences, The University of Zambia, Lusaka, 10101, Zambia
| | - Bright Chilikwazi
- Department of Chemistry, School of Natural Sciences, The University of Zambia, Lusaka, 10101, Zambia
| | - Jun Hu
- School of Chemistry and Chemical Engineering, Southeast University, Jiangning, Nanjing, Jiangsu Province, 211189, P. R. China
| | - Guodong Fu
- School of Chemistry and Chemical Engineering, Southeast University, Jiangning, Nanjing, Jiangsu Province, 211189, P. R. China
| |
Collapse
|
4
|
Lopes CF, Laurent E, Caul-Futy M, Dubois J, Mialon C, Chojnacki C, Sage E, Boda B, Huang S, Rosa-Calatrava M, Constant S. A Novel In Vitro Primary Human Alveolar Model (AlveolAir™) for H1N1 and SARS-CoV-2 Infection and Antiviral Screening. Microorganisms 2025; 13:572. [PMID: 40142465 PMCID: PMC11944821 DOI: 10.3390/microorganisms13030572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Lower respiratory infections, mostly caused by viral or bacterial pathogens, remain a leading global cause of mortality. The differences between animal models and humans contribute to inefficiencies in drug development, highlighting the need for more relevant and predictive, non-animal models. In this context, AlveolAir™, a fully primary in vitro 3D human alveolar model, was characterized and demonstrated the sustained presence of alveolar type I (ATI) and type II (ATII) cells. This model exhibited a functional barrier over a 30-day period, evidenced by high transepithelial electrical resistance (TEER). These findings were further validated by tight junctions' confocal microscopy and low permeability to Lucifer yellow, confirming AlveolAir™ as robust platform for drug transport assays. Additionally, successful infections with H1N1 and SARS-CoV-2 viruses were achieved, and antiviral treatments with Baloxavir and Remdesivir, respectively, effectively reduced viral replication. Interestingly, both viruses infected only the epithelial layer without replicating in endothelial cells. These findings indicate AlveolAir™ as a relevant model for assessing the toxicity and permeability of xenobiotics and evaluating the efficacy of novel antiviral therapies.
Collapse
Affiliation(s)
| | - Emilie Laurent
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Inserm, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France; (E.L.); (J.D.); (C.M.); (M.R.-C.)
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Research Laboratory RespiVir France-Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec Université Claude Bernard Lyon 1, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Mireille Caul-Futy
- Epithelix,1228 Geneva, Switzerland; (C.F.L.); (M.C.-F.); (C.C.); (B.B.); (S.H.)
| | - Julia Dubois
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Inserm, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France; (E.L.); (J.D.); (C.M.); (M.R.-C.)
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Research Laboratory RespiVir France-Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec Université Claude Bernard Lyon 1, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Chloé Mialon
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Inserm, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France; (E.L.); (J.D.); (C.M.); (M.R.-C.)
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Research Laboratory RespiVir France-Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec Université Claude Bernard Lyon 1, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
| | - Caroline Chojnacki
- Epithelix,1228 Geneva, Switzerland; (C.F.L.); (M.C.-F.); (C.C.); (B.B.); (S.H.)
| | | | - Bernadett Boda
- Epithelix,1228 Geneva, Switzerland; (C.F.L.); (M.C.-F.); (C.C.); (B.B.); (S.H.)
| | - Song Huang
- Epithelix,1228 Geneva, Switzerland; (C.F.L.); (M.C.-F.); (C.C.); (B.B.); (S.H.)
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Inserm, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France; (E.L.); (J.D.); (C.M.); (M.R.-C.)
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, 69008 Lyon, France
- International Research Laboratory RespiVir France-Canada, Centre de Recherche en Infectiologie, Faculté de Médecine RTH Laennec Université Claude Bernard Lyon 1, INSERM, CNRS, ENS de Lyon, 69008 Lyon, France
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Samuel Constant
- Epithelix,1228 Geneva, Switzerland; (C.F.L.); (M.C.-F.); (C.C.); (B.B.); (S.H.)
| |
Collapse
|
5
|
Qiu Y, Hu G. Lung-on-a-chip: From design principles to disease applications. BIOMICROFLUIDICS 2025; 19:021501. [PMID: 40161998 PMCID: PMC11954643 DOI: 10.1063/5.0257908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/12/2025] [Indexed: 04/02/2025]
Abstract
To address the growing need for accurate lung models, particularly in light of respiratory diseases, lung cancer, and the COVID-19 pandemic, lung-on-a-chip technology is emerging as a powerful alternative. Lung-on-a-chip devices utilize microfluidics to create three-dimensional models that closely mimic key physiological features of the human lung, such as the air-liquid interface, mechanical forces associated with respiration, and fluid dynamics. This review provides a comprehensive overview of the fundamental components of lung-on-a-chip systems, the diverse fabrication methods used to construct these complex models, and a summary of their wide range of applications in disease modeling and aerosol deposition studies. Despite existing challenges, lung-on-a-chip models hold immense potential for advancing personalized medicine, drug development, and disease prevention, offering a transformative approach to respiratory health research.
Collapse
Affiliation(s)
- Yan Qiu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Guoqing Hu
- Department of Engineering Mechanics, State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
6
|
Fuglsang-Madsen A, Haagensen JAJ, De Rudder C, Simões FB, Molin S, Johansen HK. Establishment of a 3D-Printed Tissue-on-a-Chip Model for Live Imaging of Bacterial Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 39825043 DOI: 10.1007/5584_2024_829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Despite advances in healthcare, bacterial pathogens remain a severe global health threat, exacerbated by rising antibiotic resistance. Lower respiratory tract infections, with their high death toll, are of particular concern. Accurately replicating host-pathogen interactions in laboratory models is crucial for understanding these diseases and evaluating new therapies. In this communication, we briefly present existing in vivo models for cystic fibrosis and their limitations in replicating human respiratory infections. We then present a novel, 3D-printed, cytocompatible microfluidic lung-on-a-chip device, designed to simulate the human lung environment, and with possible use in recapitulating general infectious diseases.Our device enables the colonisation of fully differentiated lung epithelia at an air-liquid interface with Pseudomonas aeruginosa, a key pathogen in many severe infections. By incorporating dynamic flow, we replicate the clearance of bacterial toxins and planktonic cells, simulating both acute and chronic infections. This platform supports real-time monitoring of therapeutic interventions, mimics repeated drug administrations as in clinical settings, and facilitates the analysis of colony-forming units and cytokine secretion over time. Our findings indicate that this lung-on-a-chip device has significant potential for advancing infectious disease research, in optimizing treatment strategies against infections and in developing novel treatments.
Collapse
Affiliation(s)
- Albert Fuglsang-Madsen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| | - Janus Anders Juul Haagensen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| | - Charlotte De Rudder
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
- Present Address: University of Luxembourg, Centre for Systems Biomedicine, Luxembourg, Belgium
| | - Filipa Bica Simões
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| | - Søren Molin
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs, Lyngby, Denmark
| | - Helle Krogh Johansen
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
G Valverde M, Stampa Zamorano C, Kožinec D, Benito Zarza L, van Genderen AM, Janssen R, Castilho M, Hrynevich A, Vermonden T, Malda J, de Ruijter M, Masereeuw R, Mihăilă SM. Thermoforming for Small Feature Replication in Melt Electrowritten Membranes to Model Kidney Proximal Tubule. Adv Healthc Mater 2025; 14:e2401800. [PMID: 39511873 DOI: 10.1002/adhm.202401800] [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: 05/15/2024] [Revised: 10/18/2024] [Indexed: 11/15/2024]
Abstract
A novel approach merging melt electrowriting (MEW) with matched die thermoforming to achieve scaffolds with micron-sized curvatures (200 - 800 µm versus 1000 µm of mandrel printing) for in vitro modeling of the kidney proximal tubule (PT) is proposed. Recent advances in this field emphasize the relevance of accurately replicating the intricate tissue microenvironment, particularly the curvature of the nephrons' tubular segments. While MEW offers promising capabilities for fabricating highly and porous precise 3D structures mimicking the PT, challenges persist in approximating the diameter of tubular scaffolds to match the actual PT. The thermoformed MEW membranes retain the initial MEW printing design parameters (rhombus geometry, porosity > 45%) while accurately following the imprinted curvature (ratios between 0.67-0.95). PT epithelial cells cultured on these membranes demonstrate the ability to fill in the large pores of the membrane by secreting their own collagen IV-rich extracellular matrix and form an organized, functional, and tight monolayer expressing characteristic PT markers. Besides approximating PT architecture, this setup maximizes the usable surface area for cell culture and molecular readouts. By closely mimicking the structural intricacies of native tissue architecture, this approach enhances the biomimetic fidelity of engineered scaffolds, offering potential applications beyond kidney tissue engineering.
Collapse
Affiliation(s)
- Marta G Valverde
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Claudia Stampa Zamorano
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Dora Kožinec
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Laura Benito Zarza
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Anne Metje van Genderen
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Robine Janssen
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Miguel Castilho
- Department of Biomedical Engineering, Technical University of Eindhoven, Eindhoven, 5612, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, Eindhoven, 513, The Netherlands
| | - Andrei Hrynevich
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, 100, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584, The Netherlands
| | - Tina Vermonden
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, Utrecht, CG 3584, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, 100, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584, The Netherlands
| | - Mylene de Ruijter
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, 100, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| | - Silvia M Mihăilă
- Department of Pharmaceutical Sciences, Div. Pharmacology, Utrecht University, Utrecht, 13102, The Netherlands
| |
Collapse
|
8
|
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
|
9
|
Fois MG, Tahmasebi Birgani ZN, López-Iglesias C, Knoops K, van Blitterswijk C, Giselbrecht S, Habibović P, Truckenmüller RK. In vitro vascularization of 3D cell aggregates in microwells with integrated vascular beds. Mater Today Bio 2024; 29:101260. [PMID: 39391792 PMCID: PMC11466645 DOI: 10.1016/j.mtbio.2024.101260] [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: 05/08/2024] [Revised: 08/20/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Most human tissues possess vascular networks supplying oxygen and nutrients. Engineering of functional tissue and organ models or equivalents often require the integration of artificial vascular networks. Several approaches, such as organs on chips and three-dimensional (3D) bioprinting, have been pursued to obtain vasculature and vascularized tissues in vitro. This technical feasibility study proposes a new approach for the in vitro vascularization of 3D microtissues. For this, we thermoform arrays of round-bottom microwells into thin non-porous and porous polymer films/membranes and culture vascular beds on them from which endothelial sprouting occurs in a Matrigel-based 3D extra cellular matrix. We present two possible culture configurations for the microwell-integrated vascular beds. In the first configuration, human umbilical vein endothelial cells (HUVECs) grow on and sprout from the inner wall of the non-porous microwells. In the second one, HUVECs grow on the outer surface of the porous microwells and sprout through the pores toward the inside. These approaches are extended to lymphatic endothelial cells. As a proof of concept, we demonstrate the in vitro vascularization of spheroids from human mesenchymal stem cells and MG-63 human osteosarcoma cells. Our results show the potential of this approach to provide the spheroids with an abundant outer vascular network and the indication of an inner vasculature.
Collapse
Affiliation(s)
- Maria G. Fois
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Zeinab N. Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, the Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
10
|
Giacomini F, Rho HS, Eischen‐Loges M, Tahmasebi Birgani Z, van Blitterswijk C, van Griensven M, Giselbrecht S, Habibović P, Truckenmüller R. Enthesitis on Chip - A Model for Studying Acute and Chronic Inflammation of the Enthesis and its Pharmacological Treatment. Adv Healthc Mater 2024; 13:e2401815. [PMID: 39188199 PMCID: PMC11650547 DOI: 10.1002/adhm.202401815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Indexed: 08/28/2024]
Abstract
Enthesitis, the inflammation of the enthesis, which is the point of attachment of tendons and ligaments to bones, is a common musculoskeletal disease. The inflammation often originates from the fibrocartilage region of the enthesis as a consequence of mechanical overuse or -load and consequently tissue damage. During enthesitis, waves of inflammatory cytokines propagate in(to) the fibrocartilage, resulting in detrimental, heterotopic bone formation. Understanding of human enthesitis and its treatment options is limited, also because of lacking in vitro model systems that can closely mimic the pathophysiology of the enthesis and can be used to develop therapies. In this study, an enthes(it)is-on-chip model is developed. On opposite sides of a porous culture membrane separating the chip's two microfluidic compartments, human mesenchymal stromal cells are selectively differentiated into tenocytes and fibrochondrocytes. By introducing an inflammatory cytokine cocktail into the fibrochondrocyte compartment, key aspects of acute and chronic enthesitis, measured as increased expression of inflammatory markers, can be recapitulated. Upon inducing chronic inflammatory conditions, hydroxyapatite deposition, enhanced osteogenic marker expression and reduced secretion of tissue-related extracellular matrix components are observed. Adding the anti-inflammatory drug celecoxib to the fibrochondrocyte compartment mitigates the inflammatory state, demonstrating the potential of the enthesitis-on-chip model for drug testing.
Collapse
Affiliation(s)
- Francesca Giacomini
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Hoon Suk Rho
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Maria Eischen‐Loges
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Clemens van Blitterswijk
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Martijn van Griensven
- Department of Cell Biology‐Inspired Tissue EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229 ERThe Netherlands
| |
Collapse
|
11
|
Ajikumar A, Lei KF. Microfluidic Technologies in Advancing Cancer Research. MICROMACHINES 2024; 15:1444. [PMID: 39770196 PMCID: PMC11677295 DOI: 10.3390/mi15121444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
This review explores the significant role of microfluidic technologies in advancing cancer research, focusing on the below key areas: droplet-based microfluidics, organ-on-chip systems, paper-based microfluidics, electrokinetic chips, and microfluidic chips for the study of immune response. Droplet-based microfluidics allows precise manipulation of cells and three-dimensional microtissues, enabling high-throughput experiments that reveal insights into cancer cell migration, invasion, and drug resistance. Organ-on-chip systems replicate human organs to assess drug efficacy and toxicity, particularly in the liver, heart, kidney, gut, lung, and brain. Paper-based microfluidics offers an alternative approach to accomplish rapid diagnostics and cell- and tissue-based bioassays. Electrokinetic microfluidic chips offer precise control over cell positioning and behavior, facilitating drug screening and cellular studies. Immune response studies leverage real-time observation of interactions between immune and cancer cells, supporting the development of immunotherapies. These microfluidic advances are paving the way for personalized cancer treatments while addressing challenges of scalability, cost, and clinical integration.
Collapse
Affiliation(s)
- Arjun Ajikumar
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Kin Fong Lei
- Department of Biomedical Engineering, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Department of Electrical & Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
12
|
Lehmann M, Krishnan R, Sucre J, Kulkarni HS, Pineda RH, Anderson C, Banovich NE, Behrsing HP, Dean CH, Haak A, Gosens R, Kaminski N, Zagorska A, Koziol-White C, Metcalf JP, Kim YH, Loebel C, Neptune E, Noel A, Raghu G, Sewald K, Sharma A, Suki B, Sperling A, Tatler A, Turner S, Rosas IO, van Ry P, Wille T, Randell SH, Pryhuber G, Rojas M, Bourke J, Königshoff M. Precision Cut Lung Slices: Emerging Tools for Preclinical and Translational Lung Research. An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 72:16-31. [PMID: 39499861 PMCID: PMC11707673 DOI: 10.1165/rcmb.2024-0479st] [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: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
The urgent need for effective treatments for acute and chronic lung diseases underscores the significance of developing innovative preclinical human research tools. The 2023 ATS Workshop on Precision Cut Lung Slices (PCLS) brought together 35 experts to discuss and address the role of human tissue-derived PCLS as a unique tool for target and drug discovery and validation in pulmonary medicine. With increasing interest and usage, along with advancements in methods and technology, there is a growing need for consensus on PCLS methodology and readouts. The current document recommends standard reporting criteria and emphasizes the requirement for careful collection and integration of clinical metadata. We further discuss current clinically relevant readouts that can be applied to PCLS and highlight recent developments and future steps for implementing novel technologies for PCLS modeling and analysis. The collection and correlation of clinical metadata and multiomic analysis will further advent the integration of this preclinical platform into patient endotyping and the development of tailored therapies for lung disease patients.
Collapse
Affiliation(s)
- Mareike Lehmann
- Philipps University Marburg, Institute for Lung Research, Marburg, Germany
- Helmholtz Center Munich, Institute for Lung Health and Immunity, Munich, Germany;
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Emergency Medicine, Boston, United States
| | - Jennifer Sucre
- Vanderbilt University Medical Center, Pediatrics, Nashville, Tennessee, United States
| | - Hrishikesh S Kulkarni
- Washington University in Saint Louis, Division of Pulmonary and Critical Care Medicine, Saint Louis, Missouri, United States
| | - Ricardo H Pineda
- University of Pittsburgh, Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | - Holger P Behrsing
- Institute for In Vitro Sciences Inc, Gaithersburg, Maryland, United States
| | - Charlotte H Dean
- Imperial College, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Haak
- Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Reinoud Gosens
- University of Groningen, Molecular Pharmacology, Groningen, Netherlands
| | - Naftali Kaminski
- Yale School of Medicine , Pulmonary, Critical Care and Sleep Mediine , New Haven, Connecticut, United States
| | - Anna Zagorska
- Gilead Sciences Inc, Foster City, California, United States
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey, United States
| | - Jordan P Metcalf
- The University of Oklahoma Health Sciences Center, Medicine, Oklahoma City, Oklahoma, United States
| | - Yong Ho Kim
- U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States
| | | | - Enid Neptune
- Johns Hopkins, Medicine/Pulmonary and Critical Care, Baltimore, Maryland, United States
| | - Alexandra Noel
- Louisiana State University, Baton Rouge, Louisiana, United States
| | - Ganesh Raghu
- University of Washington Medical Center, Division of Pulmonary and Critical Care Medicine, Seattle, Washington, United States
| | | | - Ashish Sharma
- University of Florida, Gainesville, Florida, United States
| | - Bela Suki
- Boston University, Biomedical Engineering, Boston, Massachusetts, United States
| | - Anne Sperling
- University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Amanda Tatler
- University of Nottingham, Respiratory Medicine , Nottingham, United Kingdom of Great Britain and Northern Ireland
| | - Scott Turner
- Pliant Therapeutics, South San Francisco, California, United States
| | - Ivan O Rosas
- Brigham and Women's Hospital, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Pam van Ry
- Brigham Young University, Chemistry and Biochemistry, Provo, Utah, United States
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Bundeswehr Medical Academy, Germany, Munich, Germany
| | - Scott H Randell
- University of North Carolina, Department of Cell Biology & Physiology, Chapel Hill, North Carolina, United States
| | - Gloria Pryhuber
- University of Rochester, Pediatrics, Rochester, New York, United States
| | - Mauricio Rojas
- Ohio State University, Columbus, OH, Pulmonary, Critical Care and Sleep Medicine, College of Medicine, , Columbus, Ohio, United States
| | - Jane Bourke
- Monash University, Department of Pharmacology, Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Melanie Königshoff
- University of Pittsburgh, Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
13
|
Miklosic G, Ferguson SJ, D'Este M. Engineering complex tissue-like microenvironments with biomaterials and biofabrication. Trends Biotechnol 2024; 42:1241-1257. [PMID: 38658198 DOI: 10.1016/j.tibtech.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
Advances in tissue engineering for both system modeling and organ regeneration depend on embracing and recapitulating the target tissue's functional and structural complexity. Microenvironmental features such as anisotropy, heterogeneity, and other biochemical and mechanical spatiotemporal cues are essential in regulating tissue development and function. Novel biofabrication strategies and innovative biomaterial design have emerged as promising tools to better reproduce such features. These facilitate a transition towards high-fidelity biomimetic structures, offering opportunities for a deeper understanding of tissue function and the development of superior therapies. In this review, we explore some of the key structural and compositional aspects of tissues, lay out how to achieve similar outcomes with current fabrication strategies, and identify the main challenges and promising avenues for future research.
Collapse
Affiliation(s)
- Gregor Miklosic
- AO Research Institute Davos, Davos, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
14
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A micro-scale humanized ventilator-on-a-chip to examine the injurious effects of mechanical ventilation. LAB ON A CHIP 2024; 24:4390-4402. [PMID: 39161999 PMCID: PMC11407794 DOI: 10.1039/d4lc00143e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical impedance measurements to investigate how individual and simultaneous application of mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicates that the surface tension forces associated with reopening fluid-occluded lung regions are the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to interogate the biomechanical mechanisms of VILI.
Collapse
Affiliation(s)
- Basia Gabela-Zuniga
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Vasudha C Shukla
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher Bobba
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Heather M Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, USA
- Scientific Staff, Shriners Children's Ohio, Dayton, Ohio, USA
| | - Joshua A Englert
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
15
|
Radisic M, Kaplan DL. Serving a Diverse Biomaterials Community for 10 Years. ACS Biomater Sci Eng 2024; 10:5409-5411. [PMID: 39246059 DOI: 10.1021/acsbiomaterials.4c01355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
|
16
|
Calzuola ST, Newman G, Feaugas T, Perrault CM, Blondé JB, Roy E, Porrini C, Stojanovic GM, Vidic J. Membrane-based microfluidic systems for medical and biological applications. LAB ON A CHIP 2024; 24:3579-3603. [PMID: 38954466 DOI: 10.1039/d4lc00251b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Microfluidic devices with integrated membranes that enable control of mass transport in constrained environments have shown considerable growth over the last decade. Membranes are a key component in several industrial processes such as chemical, pharmaceutical, biotechnological, food, and metallurgy separation processes as well as waste management applications, allowing for modular and compact systems. Moreover, the miniaturization of a process through microfluidic devices leads to process intensification together with reagents, waste and cost reduction, and energy and space savings. The combination of membrane technology and microfluidic devices allows therefore magnification of their respective advantages, providing more valuable solutions not only for industrial processes but also for reproducing biological processes. This review focuses on membrane-based microfluidic devices for biomedical science with an emphasis on microfluidic artificial organs and organs-on-chip. We provide the basic concepts of membrane technology and the laws governing mass transport. The role of the membrane in biomedical microfluidic devices, along with the required properties, available materials, and current challenges are summarized. We believe that the present review may be a starting point and a resource for researchers who aim to replicate a biological phenomenon on-chip by applying membrane technology, for moving forward the biomedical applications.
Collapse
Affiliation(s)
- Silvia Tea Calzuola
- UMR7646 Laboratoire d'hydrodynamique (LadHyX), Ecole Polytechnique, Palaiseau, France.
- Eden Tech, Paris, France
| | - Gwenyth Newman
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | - Thomas Feaugas
- Eden Tech, Paris, France
- Department of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Milan, Italy
| | | | | | | | | | - Goran M Stojanovic
- Faculty of Technical Sciences, University of Novi Sad, T. D. Obradovića 6, 21000 Novi Sad, Serbia
| | - Jasmina Vidic
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
17
|
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
|
18
|
Peyret C, Manousaki A, Bouguet-Bonnet S, Stratakis E, Sanchez-Gonzalez L, Kahn CJ, Arab-Tehrany E. Nanoliposomes Permeability in a Microfluidic Drug Delivery Platform across a 3D Hydrogel. Pharmaceutics 2024; 16:765. [PMID: 38931887 PMCID: PMC11207390 DOI: 10.3390/pharmaceutics16060765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Nanoliposomes are nano-sized vesicles that can be used as drug delivery carriers with the ability to encapsulate both hydrophobic and hydrophilic compounds. Moreover, their lipid compositions facilitate their internalization by cells. However, the interaction between nanoliposomes and the membrane barrier of the human body is not well-known. If cellular tests and animal testing offer a solution, their lack of physiological relevance and ethical concerns make them unsuitable to properly mimic human body complexity. Microfluidics, which allows the environment of the human body to be imitated in a controlled way, can fulfil this role. However, existing models are missing the presence of something that would mimic a basal membrane, often consisting of a simple cell layer on a polymer membrane. In this study, we investigated the diffusion of nanoliposomes in a microfluidic system and found the optimal parameters to maximize their diffusion. Then, we incorporated a custom made GelMA with a controlled degree of substitution and studied the passage of fluorescently labeled nanoliposomes through this barrier. Our results show that highly substituted GelMA was more porous than lower substitution GelMA. Overall, our work lays the foundation for the incorporation of a hydrogel mimicking a basal membrane on a drug delivery microfluidic platform.
Collapse
Affiliation(s)
- Corentin Peyret
- Université de Lorraine, LIBio, F-54000 Nancy, France; (C.P.); (L.S.-G.); (C.J.F.K.)
| | - Aleka Manousaki
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), 711 10 Heraklion, Greece; (A.M.); (E.S.)
| | | | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), 711 10 Heraklion, Greece; (A.M.); (E.S.)
| | | | - Cyril J.F. Kahn
- Université de Lorraine, LIBio, F-54000 Nancy, France; (C.P.); (L.S.-G.); (C.J.F.K.)
| | - Elmira Arab-Tehrany
- Université de Lorraine, LIBio, F-54000 Nancy, France; (C.P.); (L.S.-G.); (C.J.F.K.)
| |
Collapse
|
19
|
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
|
20
|
Carvalho DJ, Kip AM, Tegel A, Stich M, Krause C, Romitti M, Branca C, Verhoeven B, Costagliola S, Moroni L, Giselbrecht S. A Modular Microfluidic Organoid Platform Using LEGO-Like Bricks. Adv Healthc Mater 2024; 13:e2303444. [PMID: 38247306 PMCID: PMC11481080 DOI: 10.1002/adhm.202303444] [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: 10/27/2023] [Revised: 01/12/2024] [Indexed: 01/23/2024]
Abstract
The convergence of organoid and organ-on-a-chip (OoC) technologies is urgently needed to overcome limitations of current 3D in vitro models. However, integrating organoids in standard OoCs faces several technical challenges, as it is typically laborious, lacks flexibility, and often results in even more complex and less-efficient cell culture protocols. Therefore, specifically adapted and more flexible microfluidic platforms need to be developed to facilitate the incorporation of complex 3D in vitro models. Here, a modular, tubeless fluidic circuit board (FCB) coupled with reversibly sealed cell culture bricks for dynamic culture of embryonic stem cell-derived thyroid follicles is developed. The FCB is fabricated by milling channels in a polycarbonate (PC) plate followed by thermal bonding against another PC plate. LEGO-like fluidic interconnectors allow plug-and-play connection between a variety of cell culture bricks and the FCB. Lock-and-play clamps are integrated in the organoid brick to enable easy (un)loading of organoids. A multiplexed perfusion experiment is conducted with six FCBs, where thyroid organoids are transferred on-chip within minutes and cultured up to 10 d without losing their structure and functionality, thus validating this system as a flexible, easy-to-use platform, capable of synergistically combining organoids with advanced microfluidic platforms.
Collapse
Affiliation(s)
- Daniel J. Carvalho
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Anna M. Kip
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Andreas Tegel
- PreSens Precision Sensing GmbHAm Biopark 1193053RegensburgGermany
| | - Matthias Stich
- PreSens Precision Sensing GmbHAm Biopark 1193053RegensburgGermany
| | - Christian Krause
- PreSens Precision Sensing GmbHAm Biopark 1193053RegensburgGermany
| | - Mírian Romitti
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM)Université Libre de Bruxelles808 route de LennikAnderlecht1070Belgium
| | - Carlotta Branca
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Bart Verhoeven
- IDEE Instrument Development Engineering and Evaluation – Research EngineeringUniversiteitssingel 50Maastricht6200 MDThe Netherlands
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology (IRIBHM)Université Libre de Bruxelles808 route de LennikAnderlecht1070Belgium
| | - Lorenzo Moroni
- Department of Complex Tissue RegenerationMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials EngineeringMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastricht6229 ERThe Netherlands
| |
Collapse
|
21
|
Licciardello M, Traldi C, Cicolini M, Bertana V, Marasso SL, Cocuzza M, Tonda-Turo C, Ciardelli G. A miniaturized multicellular platform to mimic the 3D structure of the alveolar-capillary barrier. Front Bioeng Biotechnol 2024; 12:1346660. [PMID: 38646009 PMCID: PMC11026571 DOI: 10.3389/fbioe.2024.1346660] [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: 11/29/2023] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Several diseases affect the alveoli, and the efficacy of medical treatments and pharmaceutical therapies is hampered by the lack of pre-clinical models able to recreate in vitro the diseases. Microfluidic devices, mimicking the key structural and compositional features of the alveoli, offer several advantages to medium and high-throughput analysis of new candidate therapies. Here, we developed an alveolus-on-a-chip recapitulating the microanatomy of the physiological tissue by including the epithelium, the fibrous interstitial layer and the capillary endothelium. A PDMS device was obtained assembling a top layer and a bottom layer obtained by replica molding. A polycaprolactone/gelatin (PCL-Gel) electrospun membrane was included within the two layers supporting the seeding of 3 cell phenotypes. Epithelial cells were grown on a fibroblast-laden collagen hydrogel located on the top side of the PCL-Gel mats while endothelial cells were seeded on the basolateral side of the membrane. The innovative design of the microfluidic device allows to replicate both cell-cell and cell-extracellular matrix interactions according to the in vivo cell arrangement along with the establishment of physiologically relevant air-liquid interface conditions. Indeed, high cell viability was confirmed for up to 10 days and the formation of a tight endothelial and epithelial barrier was assessed by immunofluorescence assays.
Collapse
Affiliation(s)
- Michela Licciardello
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Cecilia Traldi
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Martina Cicolini
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Valentina Bertana
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Simone Luigi Marasso
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
- CNR-IMEM, National Research Council-Institute of Materials for Electronics and Magnetism, Parma, Italy
| | - Matteo Cocuzza
- ChiLab- Materials and Microsystems Laboratory, Politecnico di Torino, Department of Applied Science and Technology (DISAT), Chivasso, Italy
| | - Chiara Tonda-Turo
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
| | - Gianluca Ciardelli
- La.Di.Spe Bioengineerig, Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Turin, Italy
- PolitoBIOMed Lab, Politecnico di Torino, Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research, Italy
- CNR-IPCF, National Research Council-Institute for Chemical and Physical Processes, Pisa, Italy
| |
Collapse
|
22
|
Shah D, Dave B, Chorawala MR, Prajapati BG, Singh S, M. Elossaily G, Ansari MN, Ali N. An Insight on Microfluidic Organ-on-a-Chip Models for PM 2.5-Induced Pulmonary Complications. ACS OMEGA 2024; 9:13534-13555. [PMID: 38559954 PMCID: PMC10976395 DOI: 10.1021/acsomega.3c10271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024]
Abstract
Pulmonary diseases like asthma, chronic obstructive pulmonary disorder, lung fibrosis, and lung cancer pose a significant burden to global human health. Many of these complications arise as a result of exposure to particulate matter (PM), which has been examined in several preclinical and clinical trials for its effect on several respiratory diseases. Particulate matter of size less than 2.5 μm (PM2.5) has been known to inflict unforeseen repercussions, although data from epidemiological studies to back this are pending. Conventionally utilized two-dimensional (2D) cell culture and preclinical animal models have provided insufficient benefits in emulating the in vivo physiological and pathological pulmonary conditions. Three-dimensional (3D) structural models, including organ-on-a-chip models, have experienced a developmental upsurge in recent times. Lung-on-a-chip models have the potential to simulate the specific features of the lungs. With the advancement of technology, an emerging and advanced technique termed microfluidic organ-on-a-chip has been developed with the aim of identifying the complexity of the respiratory cellular microenvironment of the body. In the present Review, the role of lung-on-a-chip modeling in reproducing pulmonary complications has been explored, with a specific emphasis on PM2.5-induced pulmonary complications.
Collapse
Affiliation(s)
- Disha Shah
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Bhavarth Dave
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Mehul R. Chorawala
- Department
of Pharmacology and Pharmacy Practice, L.
M. College of Pharmacy Navrangpura, Ahmedabad, Gujarat 380009, India
| | - Bhupendra G. Prajapati
- Department
of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education and Research,
Ganpat University, Mehsana, Gujarat 384012, India
| | - Sudarshan Singh
- Office
of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
- Department
of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang
Mai 50200, Thailand
| | - Gehan M. Elossaily
- Department
of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Mohd Nazam Ansari
- Department
of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nemat Ali
- Department
of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Mancinelli E, Zushi N, Takuma M, Cheng Chau CC, Parpas G, Fujie T, Pensabene V. Porous Polymeric Nanofilms for Recreating the Basement Membrane in an Endothelial Barrier-on-Chip. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13006-13017. [PMID: 38414331 PMCID: PMC10941076 DOI: 10.1021/acsami.3c16134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/29/2024]
Abstract
Organs-on-chips (OoCs) support an organotypic human cell culture in vitro. Precise representation of basement membranes (BMs) is critical for mimicking physiological functions of tissue interfaces. Artificial membranes in polyester (PES) and polycarbonate (PC) commonly used in in vitro models and OoCs do not replicate the characteristics of the natural BMs, such as submicrometric thickness, selective permeability, and elasticity. This study introduces porous poly(d,l-lactic acid) (PDLLA) nanofilms for replicating BMs in in vitro models and demonstrates their integration into microfluidic chips. Using roll-to-roll gravure coating and polymer phase separation, we fabricated transparent ∼200 nm thick PDLLA films. These nanofilms are 60 times thinner and 27 times more elastic than PES membranes and show uniformly distributed pores of controlled diameter (0.4 to 1.6 μm), which favor cell compartmentalization and exchange of large water-soluble molecules. Human umbilical vein endothelial cells (HUVECs) on PDLLA nanofilms stretched across microchannels exhibited 97% viability, enhanced adhesion, and a higher proliferation rate compared to their performance on PES membranes and glass substrates. After 5 days of culture, HUVECs formed a functional barrier on suspended PDLLA nanofilms, confirmed by a more than 10-fold increase in transendothelial electrical resistance and blocked 150 kDa dextran diffusion. When integrated between two microfluidic channels and exposed to physiological shear stress, despite their ultrathin thickness, PDLLA nanofilms upheld their integrity and efficiently maintained separation of the channels. The successful formation of an adherent endothelium and the coculture of HUVECs and human astrocytes on either side of the suspended nanofilm validate it as an artificial BM for OoCs. Its submicrometric thickness guarantees intimate contact, a key feature to mimic the blood-brain barrier and to study paracrine signaling between the two cell types. In summary, porous PDLLA nanofilms hold the potential for improving the accuracy and physiological relevance of the OoC as in vitro models and drug discovery tools.
Collapse
Affiliation(s)
- Elena Mancinelli
- School
of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds LS2 9JT, United Kingdom
- Bragg
Centre for Materials Research, University
of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nanami Zushi
- School
of Life Science and Technology, Tokyo Institute
of Technology, B-50, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Megumi Takuma
- School
of Life Science and Technology, Tokyo Institute
of Technology, B-50, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Chalmers Chi Cheng Chau
- School
of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds LS2 9JT, United Kingdom
- Bragg
Centre for Materials Research, University
of Leeds, Leeds LS2 9JT, United Kingdom
- School
of Molecular and Cellular Biology and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - George Parpas
- School
of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds LS2 9JT, United Kingdom
- Bragg
Centre for Materials Research, University
of Leeds, Leeds LS2 9JT, United Kingdom
- Leeds
Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds LS2 9JT, United
Kingdom
| | - Toshinori Fujie
- School
of Life Science and Technology, Tokyo Institute
of Technology, B-50, Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
- Living Systems
Materialogy (LiSM) Research Group, International Research Frontiers
Initiative (IRFI), Tokyo Institute of Technology, R3-23, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8503, Japan
| | - Virginia Pensabene
- School
of Electronic and Electrical Engineering and Pollard Institute, University of Leeds, Leeds LS2 9JT, United Kingdom
- Bragg
Centre for Materials Research, University
of Leeds, Leeds LS2 9JT, United Kingdom
- Faculty
of Medicine and Health, Leeds Institute of Medical Research at St
James’s University Hospital, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
24
|
Ko J, Song J, Lee Y, Choi N, Kim HN. Understanding organotropism in cancer metastasis using microphysiological systems. LAB ON A CHIP 2024; 24:1542-1556. [PMID: 38192269 DOI: 10.1039/d3lc00889d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Cancer metastasis, the leading cause of cancer-related deaths, remains a complex challenge in medical science. Stephen Paget's "seed and soil theory" introduced the concept of organotropism, suggesting that metastatic success depends on specific organ microenvironments. Understanding organotropism not only offers potential for curbing metastasis but also novel treatment strategies. Microphysiological systems (MPS), especially organ-on-a-chip models, have emerged as transformative tools in this quest. These systems, blending microfluidics, biology, and engineering, grant precise control over cell interactions within organ-specific microenvironments. MPS enable real-time monitoring, morphological analysis, and protein quantification, enhancing our comprehension of cancer dynamics, including tumor migration, vascularization, and pre-metastatic niches. In this review, we explore innovative applications of MPS in investigating cancer metastasis, particularly focusing on organotropism. This interdisciplinary approach converges the field of science, engineering, and medicine, thereby illuminating a path toward groundbreaking discoveries in cancer research.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Jiyoung Song
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Yedam Lee
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
25
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A Micro-scale Humanized Ventilator-on-a-Chip to Examine the Injurious Effects of Mechanical Ventilation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582200. [PMID: 38464068 PMCID: PMC10925162 DOI: 10.1101/2024.02.26.582200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical resistance (TEER) measurements to investigate how individual and simultaneous application of the different mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicate that the surface tension forces associated with reopening fluid-occluded lung regions is the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to identify the biomechanical mechanisms of VILI.
Collapse
|
26
|
Chen Y, Li Z, Ji G, Wang S, Mo C, Ding B. Lung regeneration: diverse cell types and the therapeutic potential. MedComm (Beijing) 2024; 5:e494. [PMID: 38405059 PMCID: PMC10885188 DOI: 10.1002/mco2.494] [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: 07/30/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Lung tissue has a certain regenerative ability and triggers repair procedures after injury. Under controllable conditions, lung tissue can restore normal structure and function. Disruptions in this process can lead to respiratory system failure and even death, causing substantial medical burden. The main types of respiratory diseases are chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), and acute respiratory distress syndrome (ARDS). Multiple cells, such as lung epithelial cells, endothelial cells, fibroblasts, and immune cells, are involved in regulating the repair process after lung injury. Although the mechanism that regulates the process of lung repair has not been fully elucidated, clinical trials targeting different cells and signaling pathways have achieved some therapeutic effects in different respiratory diseases. In this review, we provide an overview of the cell type involved in the process of lung regeneration and repair, research models, and summarize molecular mechanisms involved in the regulation of lung regeneration and fibrosis. Moreover, we discuss the current clinical trials of stem cell therapy and pharmacological strategies for COPD, IPF, and ARDS treatment. This review provides a reference for further research on the molecular and cellular mechanisms of lung regeneration, drug development, and clinical trials.
Collapse
Affiliation(s)
- Yutian Chen
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Zhen Li
- The Department of Endovascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Gaili Ji
- Department of GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shaochi Wang
- Department of Translational MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| | - Bi‐Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
27
|
Purev E, Bahmed K, Kosmider B. Alveolar Organoids in Lung Disease Modeling. Biomolecules 2024; 14:115. [PMID: 38254715 PMCID: PMC10813493 DOI: 10.3390/biom14010115] [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: 07/26/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Lung organoids display a tissue-specific functional phenomenon and mimic the features of the original organ. They can reflect the properties of the cells, such as morphology, polarity, proliferation rate, gene expression, and genomic profile. Alveolar type 2 (AT2) cells have a stem cell potential in the adult lung. They produce and secrete pulmonary surfactant and proliferate to restore the epithelium after damage. Therefore, AT2 cells are used to generate alveolar organoids and can recapitulate distal lung structures. Also, AT2 cells in human-induced pluripotent stem cell (iPSC)-derived alveolospheres express surfactant proteins and other factors, indicating their application as suitable models for studying cell-cell interactions. Recently, they have been utilized to define mechanisms of disease development, such as COVID-19, lung cancer, idiopathic pulmonary fibrosis, and chronic obstructive pulmonary disease. In this review, we show lung organoid applications in various pulmonary diseases, drug screening, and personalized medicine. In addition, stem cell-based therapeutics and approaches relevant to lung repair were highlighted. We also described the signaling pathways and epigenetic regulation of lung regeneration. It is critical to identify novel regulators of alveolar organoid generations to promote lung repair in pulmonary diseases.
Collapse
Affiliation(s)
- Enkhee Purev
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
| | - Karim Bahmed
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
| | - Beata Kosmider
- Department of Microbiology, Immunology, and Inflammation, Temple University, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University, Philadelphia, PA 19140, USA
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA 19140, USA
- Department of Cardiovascular Sciences, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
28
|
Wang H, Ning X, Zhao F, Zhao H, Li D. Human organoids-on-chips for biomedical research and applications. Theranostics 2024; 14:788-818. [PMID: 38169573 PMCID: PMC10758054 DOI: 10.7150/thno.90492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024] Open
Abstract
Human organoids-on-chips (OrgOCs) are the synergism of human organoids (HOs) technology and microfluidic organs-on-chips (OOCs). OOCs can mimic extrinsic characteristics of organs, such as environmental clues of living tissue, while HOs are more amenable to biological analysis and genetic manipulation. By spatial cooperation, OrgOCs served as 3D organotypic living models allowing them to recapitulate critical tissue-specific properties and forecast human responses and outcomes. It represents a giant leap forward from the regular 2D cell monolayers and animal models in the improved human ecological niche modeling. In recent years, OrgOCs have offered potential promises for clinical studies and advanced the preclinical-to-clinical translation in medical and industrial fields. In this review, we highlight the cutting-edge achievements in OrgOCs, introduce the key features of OrgOCs architectures, and share the revolutionary applications in basic biology, disease modeling, preclinical assay and precision medicine. Furthermore, we discuss how to combine a wide range of disciplines with OrgOCs and accelerate translational applications, as well as the challenges and opportunities of OrgOCs in biomedical research and applications.
Collapse
Affiliation(s)
- Hui Wang
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Xiufan Ning
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Feng Zhao
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Hui Zhao
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Dong Li
- Department of Interventional & Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| |
Collapse
|
29
|
Joo H, Min S, Cho SW. Advanced lung organoids for respiratory system and pulmonary disease modeling. J Tissue Eng 2024; 15:20417314241232502. [PMID: 38406820 PMCID: PMC10894554 DOI: 10.1177/20417314241232502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
Amidst the recent coronavirus disease 2019 (COVID-19) pandemic, respiratory system research has made remarkable progress, particularly focusing on infectious diseases. Lung organoid, a miniaturized structure recapitulating lung tissue, has gained global attention because of its advantages over other conventional models such as two-dimensional (2D) cell models and animal models. Nevertheless, lung organoids still face limitations concerning heterogeneity, complexity, and maturity compared to the native lung tissue. To address these limitations, researchers have employed co-culture methods with various cell types including endothelial cells, mesenchymal cells, and immune cells, and incorporated bioengineering platforms such as air-liquid interfaces, microfluidic chips, and functional hydrogels. These advancements have facilitated applications of lung organoids to studies of pulmonary diseases, providing insights into disease mechanisms and potential treatments. This review introduces recent progress in the production methods of lung organoids, strategies for improving maturity, functionality, and complexity of organoids, and their application in disease modeling, including respiratory infection and pulmonary fibrosis.
Collapse
Affiliation(s)
- Hyebin Joo
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
| |
Collapse
|
30
|
She W, Shen C, Ying Y, Meng Q. Fabrication of sac-like hydrogel membranes for replicating curved tissue barriers on chips. LAB ON A CHIP 2023; 24:85-96. [PMID: 38018218 DOI: 10.1039/d3lc00807j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Current organ-on-a-chip (OOC) systems cannot mimic in vivo tissue barriers that feature curved geometries and rhythmic movement. This is due to the lack of a relevant membrane that can reproduce the natural biochemical and physical properties of a basement membrane, especially the characteristic sac-like structure possessed by multiple tissue barriers. To address this challenge, a sac-like hydrogel membrane is fabricated here using a one-step simple methodology inspired by soap bubble formation. Di-acrylated Pluronic® F127 (F127-DA) is a hydrogel that exhibits excellent mechanical properties, stably withstanding rhythmic mechanical stretching and fluid flow for at least 24 h. Using this hydrogel to make a membrane, a complex lung-on-a-chip device is successfully constructed, effectively replicating the alveolar-capillary barrier and demonstrating cellular function under physiological respiratory conditions. This membrane offers a crucial platform for replicating sac-like tissue barriers.
Collapse
Affiliation(s)
- Wenqi She
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| | - Chong Shen
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| | - Yinghua Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Qin Meng
- Key Laboratory of Biomass Chemical Engineering (Education Ministry), College of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China.
| |
Collapse
|
31
|
Vermeulen S, Knoops K, Duimel H, Parvizifard M, van Beurden D, López-Iglesias C, Giselbrecht S, Truckenmüller R, Habibović P, Tahmasebi Birgani Z. An in vitro model system based on calcium- and phosphate ion-induced hMSC spheroid mineralization. Mater Today Bio 2023; 23:100844. [PMID: 38033367 PMCID: PMC10682137 DOI: 10.1016/j.mtbio.2023.100844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
A challenge in regenerative medicine is creating the three-dimensional organic and inorganic in vitro microenvironment of bone, which would allow the study of musculoskeletal disorders and the generation of building blocks for bone regeneration. This study presents a microwell-based platform for creating spheroids of human mesenchymal stromal cells, which are then mineralized using ionic calcium and phosphate supplementation. The resulting mineralized spheroids promote an osteogenic gene expression profile through the influence of the spheroids' biophysical environment and inorganic signaling and require less calcium or phosphate to achieve mineralization compared to a monolayer culture. We found that mineralized spheroids represent an in vitro model for studying small molecule perturbations and extracellular mediated calcification. Furthermore, we demonstrate that understanding pathway signaling elicited by the spheroid environment allows mimicking these pathways in traditional monolayer culture, enabling similar rapid mineralization events. In sum, this study demonstrates the rapid generation and employment of a mineralized cell model system for regenerative medicine applications.
Collapse
Affiliation(s)
- Steven Vermeulen
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Kèvin Knoops
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Hans Duimel
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Maryam Parvizifard
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Denis van Beurden
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, M4I Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Zeinab Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
32
|
Shah DD, Raghani NR, Chorawala MR, Singh S, Prajapati BG. Harnessing three-dimensional (3D) cell culture models for pulmonary infections: State of the art and future directions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2861-2880. [PMID: 37266588 PMCID: PMC10235844 DOI: 10.1007/s00210-023-02541-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
Pulmonary infections have been a leading etiology of morbidity and mortality worldwide. Upper and lower respiratory tract infections have multifactorial causes, which include bacterial, viral, and rarely, fungal infections. Moreover, the recent emergence of SARS-CoV-2 has created havoc and imposes a huge healthcare burden. Drug and vaccine development against these pulmonary pathogens like respiratory syncytial virus, SARS-CoV-2, Mycobacteria, etc., requires a systematic set of tools for research and investigation. Currently, in vitro 2D cell culture models are widely used to emulate the in vivo physiologic environment. Although this approach holds a reasonable promise over pre-clinical animal models, it lacks the much-needed correlation to the in vivo tissue architecture, cellular organization, cell-to-cell interactions, downstream processes, and the biomechanical milieu. In view of these inadequacies, 3D cell culture models have recently acquired interest. Mammalian embryonic and induced pluripotent stem cells may display their remarkable self-organizing abilities in 3D culture, and the resulting organoids replicate important structural and functional characteristics of organs such the kidney, lung, gut, brain, and retina. 3D models range from scaffold-free systems to scaffold-based and hybrid models as well. Upsurge in organs-on-chip models for pulmonary conditions has anticipated encouraging results. Complexity and dexterity of developing 3D culture models and the lack of standardized working procedures are a few of the setbacks, which are expected to be overcome in the coming times. Herein, we have elaborated the significance and types of 3D cell culture models for scrutinizing pulmonary infections, along with the in vitro techniques, their applications, and additional systems under investigation.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Neha R Raghani
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, 384012, India.
| |
Collapse
|
33
|
Araújo-Gomes N, Zambito G, Johnbosco C, Calejo I, Leijten J, Löwik C, Karperien M, Mezzanotte L, Teixeira LM. Bioluminescence imaging on-chip platforms for non-invasive high-content bioimaging. Biosens Bioelectron 2023; 237:115510. [PMID: 37442028 DOI: 10.1016/j.bios.2023.115510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/09/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023]
Abstract
Incorporating non-invasive biosensing features in organ-on-chip models is of paramount importance for a wider implementation of these advanced in vitro microfluidic platforms. Optical biosensors, based on Bioluminescence Imaging (BLI), enable continuous, non-invasive, and in-situ imaging of cells, tissues or miniaturized organs without the drawbacks of conventional fluorescence imaging. Here, we report the first-of-its-kind integration and optimization of BLI in microfluidic chips, for non-invasive imaging of multiple biological readouts. The cell line HEK293T-GFP was engineered to express NanoLuc® luciferase under the control of a constitutive promoter and were cultured on-chip in 3D, in standard ECM-like hydrogels, to assess optimal cell detection conditions. Using real-time in-vitro dual-color microscopy, Bioluminescence (BL) and fluorescence (FL) were detectable using distinct imaging setups. Detection of the bioluminescent signals were observed at single cell resolution on-chip 20 min post-addition of Furimazine substrate and under perfusion. All hydrogels enabled BLI with higher signal-to-noise ratios as compared to fluorescence. For instance, agarose gels showed a ∼5-fold greater BL signal over background after injection of the substrate as compared to the FL signal. The use of BLI with microfluidic chip technologies opens up the potential for simultaneous in situ detection with continuous monitoring of multicolor cell reporters. Moreover, this can be achieved in a non-invasive manner. BL has great promise as a highly desirable biosensor for studying organ-on-chip platforms.
Collapse
Affiliation(s)
- Nuno Araújo-Gomes
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Castro Johnbosco
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Isabel Calejo
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Jeroen Leijten
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Clemens Löwik
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marcel Karperien
- Department of Developmental Bioengineering, Technical Medical Centre, University of Twente, Enschede, the Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Molecular Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Liliana Moreira Teixeira
- Department of Advanced Organ Bioengineering and Therapeutics, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
34
|
Cauli E, Polidoro MA, Marzorati S, Bernardi C, Rasponi M, Lleo A. Cancer-on-chip: a 3D model for the study of the tumor microenvironment. J Biol Eng 2023; 17:53. [PMID: 37592292 PMCID: PMC10436436 DOI: 10.1186/s13036-023-00372-6] [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: 05/30/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
The approval of anticancer therapeutic strategies is still slowed down by the lack of models able to faithfully reproduce in vivo cancer physiology. On one hand, the conventional in vitro models fail to recapitulate the organ and tissue structures, the fluid flows, and the mechanical stimuli characterizing the human body compartments. On the other hand, in vivo animal models cannot reproduce the typical human tumor microenvironment, essential to study cancer behavior and progression. This study reviews the cancer-on-chips as one of the most promising tools to model and investigate the tumor microenvironment and metastasis. We also described how cancer-on-chip devices have been developed and implemented to study the most common primary cancers and their metastatic sites. Pros and cons of this technology are then discussed highlighting the future challenges to close the gap between the pre-clinical and clinical studies and accelerate the approval of new anticancer therapies in humans.
Collapse
Affiliation(s)
- Elisa Cauli
- Department of Electronics, Information and Bioengineering, Politecnico Di Milano, Milan, Italy.
- Accelera Srl, Nerviano, Milan, Italy.
| | - Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Simona Marzorati
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico Di Milano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
35
|
Lenders V, Koutsoumpou X, Phan P, Soenen SJ, Allegaert K, de Vleeschouwer S, Toelen J, Zhao Z, Manshian BB. Modulation of engineered nanomaterial interactions with organ barriers for enhanced drug transport. Chem Soc Rev 2023; 52:4672-4724. [PMID: 37338993 DOI: 10.1039/d1cs00574j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
The biomedical use of nanoparticles (NPs) has been the focus of intense research for over a decade. As most NPs are explored as carriers to alter the biodistribution, pharmacokinetics and bioavailability of associated drugs, the delivery of these NPs to the tissues of interest remains an important topic. To date, the majority of NP delivery studies have used tumor models as their tool of interest, and the limitations concerning tumor targeting of systemically administered NPs have been well studied. In recent years, the focus has also shifted to other organs, each presenting their own unique delivery challenges to overcome. In this review, we discuss the recent advances in leveraging NPs to overcome four major biological barriers including the lung mucus, the gastrointestinal mucus, the placental barrier, and the blood-brain barrier. We define the specific properties of these biological barriers, discuss the challenges related to NP transport across them, and provide an overview of recent advances in the field. We discuss the strengths and shortcomings of different strategies to facilitate NP transport across the barriers and highlight some key findings that can stimulate further advances in this field.
Collapse
Affiliation(s)
- Vincent Lenders
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Xanthippi Koutsoumpou
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| | - Philana Phan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Stefaan J Soenen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Karel Allegaert
- Department of Hospital Pharmacy, Erasmus MC University Medical Center, CN Rotterdam, 3015, The Netherlands
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B3000 Leuven, Belgium
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Steven de Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Jaan Toelen
- Leuven Child and Youth Institute, KU Leuven, 3000 Leuven, Belgium
- Woman and Child, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium.
| |
Collapse
|
36
|
Zhang L, Liao W, Chen S, Chen Y, Cheng P, Lu X, Ma Y. Towards a New 3Rs Era in the construction of 3D cell culture models simulating tumor microenvironment. Front Oncol 2023; 13:1146477. [PMID: 37077835 PMCID: PMC10106600 DOI: 10.3389/fonc.2023.1146477] [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: 01/17/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Three-dimensional cell culture technology (3DCC) sits between two-dimensional cell culture (2DCC) and animal models and is widely used in oncology research. Compared to 2DCC, 3DCC allows cells to grow in a three-dimensional space, better simulating the in vivo growth environment of tumors, including hypoxia, nutrient concentration gradients, micro angiogenesis mimicism, and the interaction between tumor cells and the tumor microenvironment matrix. 3DCC has unparalleled advantages when compared to animal models, being more controllable, operable, and convenient. This review summarizes the comparison between 2DCC and 3DCC, as well as recent advances in different methods to obtain 3D models and their respective advantages and disadvantages.
Collapse
Affiliation(s)
- Long Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiqi Liao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shimin Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yukun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pengrui Cheng
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinjun Lu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Ma
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
Ramalho AS, Amato F, Gentzsch M. Patient-derived cell models for personalized medicine approaches in cystic fibrosis. J Cyst Fibros 2023; 22 Suppl 1:S32-S38. [PMID: 36529661 PMCID: PMC9992303 DOI: 10.1016/j.jcf.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) channel that perturb anion transport across the epithelia of the airways and other organs. To treat cystic fibrosis, strategies that target mutant CFTR have been developed such as correctors that rescue folding and enhance transfer of CFTR to the apical membrane, and potentiators that increase CFTR channel activity. While there has been tremendous progress in development and approval of CFTR therapeutics for the most common (F508del) and several other CFTR mutations, around 10-20% of people with cystic fibrosis have rare mutations that are still without an effective treatment. In the current decade, there was an impressive evolution of patient-derived cell models for precision medicine. In cystic fibrosis, these models have played a crucial role in characterizing the molecular defects in CFTR mutants and identifying compounds that target these defects. Cells from nasal, bronchial, and rectal epithelia are most suitable to evaluate treatments that target CFTR. In vitro assays using cultures grown at an air-liquid interface or as organoids and spheroids allow the diagnosis of the CFTR defect and assessment of potential treatment strategies. An overview of currently established cell culture models and assays for personalized medicine approaches in cystic fibrosis will be provided in this review. These models allow theratyping of rare CFTR mutations with available modulator compounds to predict clinical efficacy. Besides evaluation of individual personalized responses to CFTR therapeutics, patient-derived culture models are valuable for testing responses to developmental treatments such as novel RNA- and DNA-based therapies.
Collapse
Affiliation(s)
- Anabela S Ramalho
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Felice Amato
- Department Of Molecular Medicine and Medical Biotechnologies and CE.IN.GE - Biotecnologie Avanzate, University of Naples Federico II, Naples, Italy
| | - Martina Gentzsch
- Marsico Lung Institute - Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
38
|
Polymer film-based microwell array platform for long-term culture and research of human bronchial organoids. Mater Today Bio 2023; 19:100603. [PMID: 37009070 PMCID: PMC10060184 DOI: 10.1016/j.mtbio.2023.100603] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 03/15/2023] Open
Abstract
The culture of lung organoids relies on drops of basement membrane matrices. This comes with limitations, for example, concerning the microscopic monitoring and imaging of the organoids in the drops. Also, the culture technique is not easily compatible with micromanipulations of the organoids. In this study, we investigated the feasibility of the culture of human bronchial organoids in defined x-, y- and z-positions in a polymer film-based microwell array platform. The circular microwells have thin round/U-bottoms. For this, single cells are first precultured in drops of basement membrane extract (BME). After they form cell clusters or premature organoids, the preformed structures are then transferred into the microwells in a solution of 50% BME in medium. There, the structures can be cultured toward differentiated and mature organoids for several weeks. The organoids were characterized by bright-field microscopy for size growth and luminal fusion over time, by scanning electron microscopy for overall morphology, by transmission electron microscopy for the existence of microvilli and cilia, by video microscopy for beating cilia and swirling fluid, by live-cell imaging, by fluorescence microscopy for the expression of cell-specific markers and for proliferating and apoptotic cells, and by ATP measurement for extended cell viability. Finally, we demonstrated the eased micromanipulation of the organoids in the microwells by the example of their microinjection.
Collapse
|
39
|
Doryab A, Groll J. Biomimetic In Vitro Lung Models: Current Challenges and Future Perspective. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210519. [PMID: 36750972 DOI: 10.1002/adma.202210519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/10/2022] [Indexed: 06/18/2023]
Abstract
As post-COVID complications, chronic respiratory diseases are one of the foremost causes of mortality. The quest for a cure for this recent global challenge underlines that the lack of predictive in vitro lung models is one of the main bottlenecks in pulmonary preclinical drug development. Despite rigorous efforts to develop biomimetic in vitro lung models, the current cutting-edge models represent a compromise in numerous technological and biological aspects. Most advanced in vitro models are still in the "proof-of-concept" phase with a low clinical translation of the findings. On the other hand, advances in cellular and molecular studies are mainly based on relatively simple and unrealistic in vitro models. Herein, the current challenges and potential strategies toward not only bioinspired but truly biomimetic lung models are discussed.
Collapse
Affiliation(s)
- Ali Doryab
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Neuherberg, 85764, Munich, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), and Bavarian Polymer Institute (BPI), University of Würzburg, 97070, Würzburg, Germany
| |
Collapse
|
40
|
Wang H, Yin F, Li Z, Su W, Li D. Advances of microfluidic lung chips for assessing atmospheric pollutants exposure. ENVIRONMENT INTERNATIONAL 2023; 172:107801. [PMID: 36774736 DOI: 10.1016/j.envint.2023.107801] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Atmospheric pollutants, including particulate matters, nanoparticles, bioaerosols, and some chemicals, have posed serious threats to the environment and the human's health. The lungs are the responsible organs for providing the interface betweenthecirculatory system and the external environment, where pollutant particles can deposit or penetrate into bloodstream circulation. Conventional studies to decipher the mechanismunderlying air pollution and human health are quite limited, due to the lack of reliable models that can reproduce in vivo features of lung tissues after pollutants exposure. In the past decade, advanced near-to-native lung chips, combining cell biology with bioengineered technology, present a new strategy for atmospheric pollutants assessment and narrow the gap between 2D cell culture and in vivo animal models. In this review, the key features of artificial lung chips and the cutting-edge technologies of the lung chip manufacture are introduced. The recent progresses of lung chip technologies for atmospheric pollutants exposure assessment are summarized and highlighted. We further discuss the current challenges and the future opportunities of the development of advanced lung chips and their potential utilities in atmospheric pollutants associated toxicity testing and drug screening.
Collapse
Affiliation(s)
- Hui Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fangchao Yin
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Zhongyu Li
- College of Life Science, Dalian Minzu University, Dalian 116600, China
| | - Wentao Su
- Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034 Liaoning, China.
| | - Dong Li
- Medical School, Nantong University, Nantong 226001, China.
| |
Collapse
|
41
|
Ni K, Che B, Yang C, Qin Y, Gu R, Wang C, Luo M, Deng L. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Front Pharmacol 2022; 13:1033043. [PMID: 36578545 PMCID: PMC9790924 DOI: 10.3389/fphar.2022.1033043] [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: 08/31/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a never before seen challenge to human health and the world economy. However, it is difficult to widely use conventional animal and cell culture models in understanding the underlying pathological mechanisms of COVID-19, which in turn hinders the development of relevant therapeutic treatments, including drugs. To overcome this challenge, various three-dimensional (3D) pulmonary cell culture models such as organoids are emerging as an innovative toolset for simulating the pathophysiology occurring in the respiratory system, including bronchial airways, alveoli, capillary network, and pulmonary interstitium, which provide a robust and powerful platform for studying the process and underlying mechanisms of SARS-CoV-2 infection among the potential primary targets in the lung. This review introduces the key features of some of these recently developed tools, including organoid, lung-on-a-chip, and 3D bioprinting, which can recapitulate different structural compartments of the lung and lung function, in particular, accurately resembling the human-relevant pathophysiology of SARS-CoV-2 infection in vivo. In addition, the recent progress in developing organoids for alveolar and airway disease modeling and their applications for discovering drugs against SARS-CoV-2 infection are highlighted. These innovative 3D cell culture models together may hold the promise to fully understand the pathogenesis and eventually eradicate the pandemic of COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| |
Collapse
|
42
|
Lee DF, Thompson CL, Baynes RE, Enomoto H, Smith GW, Chambers MA. Development and evaluation of a bovine lung-on-chip (bLOC) to study bovine respiratory diseases. IN VITRO MODELS 2022; 1:333-346. [PMID: 36660607 PMCID: PMC9383688 DOI: 10.1007/s44164-022-00030-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/22/2023]
Abstract
Purpose Current air-liquid interface (ALI) models of bovine proximal airways have their limitations. They do not simulate blood flow necessary to mimic systemic drug administration, and repeated sampling requires multiple, independent cultures. A bovine lung-on-chip (bLOC) would overcome these limitations, providing a convenient and cost-effective model for pharmacokinetic or pathogenicity studies. Methods Bovine pulmonary arterial endothelial cells seeded into the endothelial channel of an Emulate Lung-Chip were interfaced with bovine bronchial epithelial cells in the epithelial channel. Cells were cultured at ALI for up to 21 days. Differentiation was assessed by mucin quantification, phase-contrast light microscopy and immunofluorescence of cell-specific markers in fixed cultures. Barrier integrity was determined by FITC-labelled dextran 3-5 kDa permeability. To evaluate the model, endothelial-epithelial transport of the antibiotic drug, danofloxacin, was followed using liquid chromatography-mass spectrometry, with the aim of replicating data previously determined in vivo. Results bLOC cultures secreted quantifiable mucins, whilst cilia formation was evident in the epithelial channel. Barrier integrity of the model was demonstrated by resistance to FITC-Dextran 3-5 kDa permeation. Bronchial epithelial and endothelial cell-specific markers were observed. Close to plasma, representative PK data for danofloxacin was observed in the endothelial channel; however, danofloxacin in the epithelial channel was mostly below the limit of quantification. Conclusion A co-culture model of the bovine proximal airway was successfully generated, with potential to replace in vivo experimentation. With further optimisation and characterisation, the bLOC may be suitable to perform drug pharmacokinetic studies for bovine respiratory disease (BRD), and other applications.
Collapse
Affiliation(s)
- Diane F. Lee
- School of Veterinary Medicine, University of Surrey, Guildford, UK
- Now at Sussex Drug Discovery Centre, University of Sussex, Falmer, UK
| | - Clare L. Thompson
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Ronald E. Baynes
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | - Hiroko Enomoto
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC USA
| | | | - Mark A. Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, UK
| |
Collapse
|