1
|
Capandova M, Sedlakova V, Vorac Z, Kotasova H, Dumkova J, Moran L, Jaros J, Antol M, Bohaciakova D, Hampl A. Using Polycaprolactone Nanofibers for the Proof-of-Concept Construction of the Alveolar-Capillary Interface. J Biomed Mater Res A 2025; 113:e37824. [PMID: 39474705 DOI: 10.1002/jbm.a.37824] [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: 05/29/2024] [Revised: 09/14/2024] [Accepted: 10/14/2024] [Indexed: 12/26/2024]
Abstract
The alveolar-capillary interface is the key functional element of gas exchange in the human lung, and disruptions to this interface can lead to significant medical complications. However, it is currently challenging to adequately model this interface in vitro, as it requires not only the co-culture of human alveolar epithelial and endothelial cells but mainly the preparation of a biocompatible scaffold that mimics the basement membrane. This scaffold should support cell seeding from both sides, and maintain optimal cell adhesion, growth, and differentiation conditions. Our study investigates the use of polycaprolactone (PCL) nanofibers as a versatile substrate for such cell cultures, aiming to model the alveolar-capillary interface more accurately. We optimized nanofiber production parameters, utilized polyamide mesh UHELON as a mechanical support for scaffold handling, and created 3D-printed inserts for specialized co-cultures. Our findings confirm that PCL nanofibrous scaffolds are manageable and support the co-culture of diverse cell types, effectively enabling cell attachment, proliferation, and differentiation. Our research establishes a proof-of-concept model for the alveolar-capillary interface, offering significant potential for enhancing cell-based testing and advancing tissue-engineering applications that require specific nanofibrous matrices.
Collapse
Affiliation(s)
- Michaela Capandova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Computer Science, Masaryk University, Brno, Czech Republic
| | - Veronika Sedlakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zbynek Vorac
- Department of Plasma Physics and Technology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Hana Kotasova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Dumkova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lukas Moran
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Josef Jaros
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Matej Antol
- Institute of Computer Science, Masaryk University, Brno, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, Czech Republic
| | - Ales Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital in Brno, Brno, Czech Republic
| |
Collapse
|
2
|
Iriondo C, Koornneef S, Skarp KP, Buscop-van Kempen M, Boerema-de Munck A, Rottier RJ. Simple-Flow: A 3D-Printed Multiwell Flow Plate to Coculture Primary Human Lung Cells at the Air-Liquid Interface. ACS Biomater Sci Eng 2024. [PMID: 39719361 DOI: 10.1021/acsbiomaterials.4c01322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Immortalized epithelial cell lines and animal models have been used in fundamental and preclinical research to study pulmonary diseases. However valuable, though, these models incompletely recapitulate the in vivo human lung, which leads to low predictive outcomes in potential respiratory treatments. Advanced technology and cell culture techniques stimulate the development of improved models that more closely mimic the physiology of the human lung. Nonetheless, most of these models are technically demanding and have a low throughput and reproducibility. Here, we describe a robust fluidic device consisting of a biocompatible and customizable 3D-printed cell culture plate, the Simple-Flow, which has medium throughput, is simple to manufacture, and is easy to set up. As a proof of principle, human primary bronchial epithelial cells (hPBECs) and human pulmonary microvascular endothelial cells (hMVECs) were cocultured on the apical and basolateral sides of the inset membranes, respectively. While hPBECs were cultured at the air-liquid interface to induce mucociliary differentiation, hMVECs were exposed to flow medium for up to 2 weeks. We show the versatility of 3D-printing technology in designing in vitro models for cell culturing applications, such as pediatric lung diseases or other pulmonary disorders.
Collapse
Affiliation(s)
- Cinta Iriondo
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Sem Koornneef
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Kari-Pekka Skarp
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Marjon Buscop-van Kempen
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Anne Boerema-de Munck
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Sophia Children's Hospital, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CB, The Netherlands
| |
Collapse
|
3
|
Yang X, Shi J, Shi B, Li J, Xue C, Ma J, Gao X. Micro- and nano-fibers for organ-on-a-chip: Construction, applications, and prospects. Mater Today Bio 2024; 29:101322. [PMID: 39554843 PMCID: PMC11567939 DOI: 10.1016/j.mtbio.2024.101322] [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: 08/22/2024] [Revised: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Organ-on-a-chip, an in vitro biomimetic microsystem that enables precise regulation and real-time observation of the cell microenvironment, has the potential to become a powerful platform for recapitulating the real microenvironment of organs in vitro. Microenvironmental factors, such as living cells, three-dimensional (3D) culture, tissue-tissue interfaces, and biomechanical factors, are important cues in the construction of biomimetic microsystems. It is important to provide an appropriate 3D culture environment for living cells to grow. Fibers, particularly microfibers and nanofibers, can provide a suitable 3D culture environment for living cells via surface adhesion or internal loading. In addition, fibers can further expand their applications in tissue engineering and biomedical research by being assembled at a higher level in various ways to create functional 3D tissues or organs with more complex structures. The use of fiber to construct an organ-on-a-chip, whether as a 3D scaffold for cell culture or to more closely mimic real tissues/organs, will introduce new ideas and strategies for developing novel organ-on-a-chip systems. Based on this context, this review summarizes the research progress in the construction and applications of micro/nanofibers for organ-on-a-chip systems. It outlines the preparation methods and material selections for micro/nanofibers and provides a detailed overview of their respective strategies for cell 3D culture and organ-on-a-chip construction. This review also highlights the main research findings and applications of micro/nanofiber in this field, which have significant implications for future practice, and finally concludes by examining potential directions for future development.
Collapse
Affiliation(s)
- Xiaoling Yang
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| | - Jingyan Shi
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| | - Bori Shi
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| | - Jianing Li
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| | - Chang Xue
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| | - Jingyun Ma
- Ningbo Institute of Innovation for Combined Medicine and Engineering, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
4
|
Filiz Y, Esposito A, De Maria C, Vozzi G, Yesil-Celiktas O. A comprehensive review on organ-on-chips as powerful preclinical models to study tissue barriers. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:042001. [PMID: 39655848 DOI: 10.1088/2516-1091/ad776c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 09/04/2024] [Indexed: 12/18/2024]
Abstract
In the preclinical stage of drug development, 2D and 3D cell cultures under static conditions followed by animal models are utilized. However, these models are insufficient to recapitulate the complexity of human physiology. With the developing organ-on-chip (OoC) technology in recent years, human physiology and pathophysiology can be modeled better than traditional models. In this review, the need for OoC platforms is discussed and evaluated from both biological and engineering perspectives. The cellular and extracellular matrix components are discussed from a biological perspective, whereas the technical aspects such as the intricate working principles of these systems, the pivotal role played by flow dynamics and sensor integration within OoCs are elucidated from an engineering perspective. Combining these two perspectives, bioengineering applications are critically discussed with a focus on tissue barriers such as blood-brain barrier, ocular barrier, nasal barrier, pulmonary barrier and gastrointestinal barrier, featuring recent examples from the literature. Furthermore, this review offers insights into the practical utility of OoC platforms for modeling tissue barriers, showcasing their potential and drawbacks while providing future projections for innovative technologies.
Collapse
Affiliation(s)
- Yagmur Filiz
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 8500 Kortrijk, Belgium
| | - Alessio Esposito
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- EgeSAM-Ege University Translational Pulmonary Research Center, Bornova, Izmir, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| |
Collapse
|
5
|
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
|
6
|
Janssen R, Benito-Zarza L, Cleijpool P, Valverde MG, Mihăilă SM, Bastiaan-Net S, Garssen J, Willemsen LEM, Masereeuw R. Biofabrication Directions in Recapitulating the Immune System-on-a-Chip. Adv Healthc Mater 2024:e2304569. [PMID: 38625078 DOI: 10.1002/adhm.202304569] [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: 12/21/2023] [Revised: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Ever since the implementation of microfluidics in the biomedical field, in vitro models have experienced unprecedented progress that has led to a new generation of highly complex miniaturized cell culture platforms, known as Organs-on-a-Chip (OoC). These devices aim to emulate biologically relevant environments, encompassing perfusion and other mechanical and/or biochemical stimuli, to recapitulate key physiological events. While OoCs excel in simulating diverse organ functions, the integration of the immune organs and immune cells, though recent and challenging, is pivotal for a more comprehensive representation of human physiology. This comprehensive review covers the state of the art in the intricate landscape of immune OoC models, shedding light on the pivotal role of biofabrication technologies in bridging the gap between conceptual design and physiological relevance. The multifaceted aspects of immune cell behavior, crosstalk, and immune responses that are aimed to be replicated within microfluidic environments, emphasizing the need for precise biomimicry are explored. Furthermore, the latest breakthroughs and challenges of biofabrication technologies in immune OoC platforms are described, guiding researchers toward a deeper understanding of immune physiology and the development of more accurate and human predictive models for a.o., immune-related disorders, immune development, immune programming, and immune regulation.
Collapse
Affiliation(s)
- Robine Janssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Laura Benito-Zarza
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Pim Cleijpool
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Marta G Valverde
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Silvia M Mihăilă
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, 6708 WG, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Danone Global Research & Innovation Center, Danone Nutricia Research B.V., Utrecht, 3584 CT, The Netherlands
| | - Linette E M Willemsen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Sen RK, Prabhakar P, Shruti, Verma P, Vikram A, Mishra A, Dwivedi A, Gowri VS, Chaurasia JP, Mondal DP, Srivastava AK, Dwivedi N, Dhand C. Smart Nanofibrous Hydrogel Wound Dressings for Dynamic Infection Diagnosis and Control: Soft but Functionally Rigid. ACS APPLIED BIO MATERIALS 2024; 7:999-1016. [PMID: 38198289 DOI: 10.1021/acsabm.3c01000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Chronic wounds, such as burns and diabetic foot ulcers, pose significant challenges to global healthcare systems due to prolonged hospitalization and increased costs attributed to susceptibility to bacterial infections. The conventional use of antibiotic-loaded and metal-impregnated dressings exacerbates concerns related to multidrug resistance and skin argyrosis. In response to these challenges, our research introduces a unique approach utilizing antibiotic-free smart hydrogel wound dressings with integrated infection eradication and diagnostic capabilities. Electrospinning stands out as a method capable of producing hydrogel nanofibrous materials possessing favorable characteristics for treating wounds and detecting infections under conditions utilizing sustainable materials. In this study, innovative dressings are fabricated through electrospinning polycaprolactone (PCL)/gelatin (GEL) hybrid hydrogel nanofibers, incorporating pDA as a cross-linker, εPL as a broad-spectrum antimicrobial agent, and anthocyanin as a pH-responsive probe. The developed dressings demonstrate exceptional antioxidant (>90% radical scavenging) and antimicrobial properties (95-100% killing). The inclusion of polyphenols/flavonoids and εPL leads to absolute bacterial eradication, and in vitro assessments using HaCaT cells indicate increased cell proliferation, decreased reactive oxygen species (ROS) production, and enhanced cell viability (100% Cell viability). The dressings display notable alterations in color that correspond to different wound conditions. Specifically, they exhibit a red/violet hue under healthy wound conditions (pH 4-6.5) and a green/blue color under unhealthy wound conditions (pH > 6.5). These distinctive color changes provide valuable insights into the versatile applications of the dressings in the care and management of wounds. Our findings suggest that these antibiotic-free smart hydrogel wound dressings hold promise as an effective and sustainable solution for chronic wounds, providing simultaneous infection control and diagnostic monitoring. This research contributes to advancing the field of wound care, offering a potential paradigm shift in the development of next-generation wound dressings.
Collapse
Affiliation(s)
- Raj Kumar Sen
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priyanka Prabhakar
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shruti
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priya Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- CSIR-National Botanical Research Institute, Rana Pratap Marg, Lucknow 226001, India
| | - Apeksha Vikram
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Aradhana Mishra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- CSIR-National Botanical Research Institute, Rana Pratap Marg, Lucknow 226001, India
| | - Ashish Dwivedi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Vijay Sorna Gowri
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jamuna Prasad Chaurasia
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dehi Pada Mondal
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Avanish Kumar Srivastava
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeraj Dwivedi
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chetna Dhand
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal 462026, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
11
|
Licciardello M, Sgarminato V, Ciardelli G, Tonda-Turo C. Development of biomimetic co-culture and tri-culture models to mimic the complex structure of the alveolar-capillary barrier. BIOMATERIALS ADVANCES 2023; 154:213620. [PMID: 37690344 DOI: 10.1016/j.bioadv.2023.213620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/29/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Alveoli are the functional area of respiratory system where the gaseous exchanges take place at level of the alveolar-capillary barrier. The development of safe and effective therapeutic approaches for treating lung disease is currently limited due to the lack of realistic preclinical models for their testing and validation. In this work, tissue engineering approaches were exploited to develop a biomimetic platform that provide an appropriate mimicking of the extracellular environment and the multicellular architecture of human alveoli. Here, we propose the implementation of two biomimetic in vitro models to reproduce the features of the main anatomic portions of the physiological alveolar-capillary barrier. First, a co-culture barrier model was obtained by integrating an electrospun polycaprolactone-gelatin (PCL-Gel) membrane in a modified transwell insert (PCL-Gel TW) to mimic the alveolar basement membrane (coded as thin model). Alveolar epithelial (A549) and lung microvascular endothelial (HULEC-5a) cells were cultured on the apical and basolateral side of the PCL-Gel membrane, respectively, under physiologic air-liquid interface (ALI) conditions for 7 days. The ALI condition promoted the expression of type I and type II alveolar epithelial cell markers and the secretion of mucus in A549 cells. Increased cell viability and barrier properties in co-cultures of A549 and HULEC-5a compared to mono-cultures revealed the effectiveness of the model to reproduce in vitro physiological-relevant features of the alveolar-capillary barrier. The second portion of the alveolar-capillary barrier was developed implementing a tri-culture model (coded as thick model) including a type I collagen (COLL) hydrogel formulated to host lung fibroblasts (MRC-5). The thick barrier model was implemented by seeding HULEC-5a on the basolateral side of PCL-Gel TW and then pouring sequentially MRC-5-laden COLL hydrogel and A549 cells on the apical side of the electrospun membrane. The thick model was maintained up to 7 days at ALI and immunofluorescence staining of tight and adherent junctions demonstrated the formation of a tight barrier. Lastly, the ability of models to emulate pathological inflammatory conditions was validated by exposing the apical compartment of the PCL-Gel TW to lipopolysaccharide (LPS). The damage of A549 tight junctions, the increase of barrier permeability and IL-6 pro-inflammatory cytokine release was observed after 48 h exposure to LPS.
Collapse
Affiliation(s)
- Michela Licciardello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy
| | - Viola Sgarminato
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, 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
| | - Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; POLITO BIOMedLAB, Politecnico di Torino, Turin, Italy; Interuniversity Center for the promotion of the 3Rs principles in teaching and research, Italy.
| |
Collapse
|
12
|
Man K, Liu J, Liang C, Corona C, Story MD, Meckes B, Yang Y. Biomimetic Human Lung Alveolar Interstitium Chip with Extended Longevity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:36888-36898. [PMID: 37463843 DOI: 10.1021/acsami.3c04091] [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] [Indexed: 07/20/2023]
Abstract
Determining the mechanistic causes of lung diseases, developing new treatments thereof, and assessing toxicity whether from chemical exposures or engineered nanomaterials would benefit significantly from a preclinical human lung alveolar interstitium model of physiological relevance. The existing preclinical models have limitations because they fail to replicate the key anatomical and physiological characteristics of human alveoli. Thus, a human lung alveolar interstitium chip was developed to imitate key alveolar microenvironmental factors including an electrospun nanofibrous membrane as the analogue of the basement membrane for co-culture of epithelial cells with fibroblasts embedded in 3D collagenous gels, physiologically relevant interstitial matrix stiffness, interstitial fluid flow, and 3D breathing-like mechanical stretch. The biomimetic chip substantially improved the epithelial barrier function compared to transwell models. Moreover, the chip having a gel made of a collagen I-fibrin blend as the interstitial matrix sustained the interstitium integrity and further enhanced the epithelial barrier, resulting in a longevity that extended beyond eight weeks. The assessment of multiwalled carbon nanotube toxicity on the chip was in line with the animal study.
Collapse
Affiliation(s)
- Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Cindy Liang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Christopher Corona
- Anne Burnett Marion School of Medicine, Texas Christian University, Fort Worth, Texas 76129, United States
| | - Michael D Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
13
|
Jain P, Rimal R, Möller M, Singh S. Topographical influence of electrospun basement membrane mimics on formation of cellular monolayer. Sci Rep 2023; 13:8382. [PMID: 37225757 DOI: 10.1038/s41598-023-34934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
Functional unit of many organs like lung, kidney, intestine, and eye have their endothelial and epithelial monolayers physically separated by a specialized extracellular matrix called the basement membrane. The intricate and complex topography of this matrix influences cell function, behavior and overall homeostasis. In vitro barrier function replication of such organs requires mimicking of these native features on an artificial scaffold system. Apart from chemical and mechanical features, the choice of nano-scale topography of the artificial scaffold is integral, however its influence on monolayer barrier formation is unclear. Though studies have reported improved single cell adhesion and proliferation in presence of pores or pitted topology, corresponding influence on confluent monolayer formation is not well reported. In this work, basement membrane mimic with secondary topographical cues is developed and its influence on single cells and their monolayers is investigated. We show that single cells cultured on fibers with secondary cues form stronger focal adhesions and undergo increased proliferation. Counterintuitively, absence of secondary cues promoted stronger cell-cell interaction in endothelial monolayers and promoted formation of integral tight barriers in alveolar epithelial monolayers. Overall, this work highlights the importance of choice of scaffold topology to develop basement barrier function in in vitro models.
Collapse
Affiliation(s)
- Puja Jain
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| | - Martin Möller
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Smriti Singh
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany.
| |
Collapse
|
14
|
Rothen-Rutishauser B, Gibb M, He R, Petri-Fink A, Sayes CM. Human lung cell models to study aerosol delivery - considerations for model design and development. Eur J Pharm Sci 2023; 180:106337. [PMID: 36410570 DOI: 10.1016/j.ejps.2022.106337] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Human lung tissue models range from simple monolayer cultures to more advanced three-dimensional co-cultures. Each model system can address the interactions of different types of aerosols and the choice of the model and the mode of aerosol exposure depends on the relevant scenario, such as adverse outcomes and endpoints of interest. This review focuses on the functional, as well as structural, aspects of lung tissue from the upper airway to the distal alveolar compartments as this information is relevant for the design of a model as well as how the aerosol properties determine the interfacial properties with the respiratory wall. The most important aspects on how to design lung models are summarized with a focus on (i) choice of appropriate scaffold, (ii) selection of cell types for healthy and diseased lung models, (iii) use of culture condition and assembly, (iv) aerosol exposure methods, and (v) endpoints and verification process. Finally, remaining challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Barbara Rothen-Rutishauser
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland.
| | - Matthew Gibb
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA
| | - Ruiwen He
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA.
| |
Collapse
|
15
|
Zamprogno P, Schulte J, Ferrari D, Rechberger K, Sengupta A, van Os L, Weber T, Zeinali S, Geiser T, Guenat OT. Lung-on-a-Chip Models of the Lung Parenchyma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:191-211. [PMID: 37195532 DOI: 10.1007/978-3-031-26625-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Since the publication of the first lung-on-a-chip in 2010, research has made tremendous progress in mimicking the cellular environment of healthy and diseased alveoli. As the first lung-on-a-chip products have recently reached the market, innovative solutions to even better mimic the alveolar barrier are paving the way for the next generation lung-on-chips. The original polymeric membranes made of PDMS are being replaced by hydrogel membranes made of proteins from the lung extracellular matrix, whose chemical and physical properties exceed those of the original membranes. Other aspects of the alveolar environment are replicated, such as the size of the alveoli, their three-dimensional structure, and their arrangement. By tuning the properties of this environment, the phenotype of alveolar cells can be tuned, and the functions of the air-blood barrier can be reproduced, allowing complex biological processes to be mimicked. Lung-on-a-chip technologies also provide the possibility of obtaining biological information that was not possible with conventional in vitro systems. Pulmonary edema leaking through a damaged alveolar barrier and barrier stiffening due to excessive accumulation of extracellular matrix proteins can now be reproduced. Provided that the challenges of this young technology are overcome, there is no doubt that many application areas will benefit greatly.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Jan Schulte
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Karin Rechberger
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Lisette van Os
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Tobias Weber
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Soheila Zeinali
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland.
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Doryab A, Taskin MB, Stahlhut P, Groll J, Schmid O. Real-Time Measurement of Cell Mechanics as a Clinically Relevant Readout of an In Vitro Lung Fibrosis Model Established on a Bioinspired Basement Membrane. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2205083. [PMID: 36030365 DOI: 10.1002/adma.202205083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Lung fibrosis, one of the major post-COVID complications, is a progressive and ultimately fatal disease without a cure. Here, an organ- and disease-specific in vitro mini-lung fibrosis model equipped with noninvasive real-time monitoring of cell mechanics is introduced as a functional readout. To establish an intricate multiculture model under physiologic conditions, a biomimetic ultrathin basement (biphasic elastic thin for air-liquid culture conditions, BETA) membrane (<1 µm) is developed with unique properties, including biocompatibility, permeability, and high elasticity (<10 kPa) for cell culturing under air-liquid interface and cyclic mechanical stretch conditions. The human-based triple coculture fibrosis model, which includes epithelial and endothelial cell lines combined with primary fibroblasts from idiopathic pulmonary fibrosis patients established on the BETA membrane, is integrated into a millifluidic bioreactor system (cyclic in vitro cell-stretch, CIVIC) with dose-controlled aerosolized drug delivery, mimicking inhalation therapy. The real-time measurement of cell/tissue stiffness (and compliance) is shown as a clinical biomarker of the progression/attenuation of fibrosis upon drug treatment, which is confirmed for inhaled Nintedanib-an antifibrosis drug. The mini-lung fibrosis model allows the combined longitudinal testing of pharmacodynamics and pharmacokinetics of drugs, which is expected to enhance the predictive capacity of preclinical models and hence facilitate the development of approved therapies for lung fibrosis.
Collapse
Affiliation(s)
- Ali Doryab
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Neuherberg, Germany
- Comprehensive Pneumology Center-Munich (CPC-M) bioArchive, Helmholtz Munich, 81377, Munich, Germany
| | - Mehmet Berat Taskin
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, 97070, Würzburg, Germany
| | - Philipp Stahlhut
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, 97070, Würzburg, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute (BPI), University of Würzburg, 97070, Würzburg, Germany
| | - Otmar Schmid
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Neuherberg, Germany
- Comprehensive Pneumology Center-Munich (CPC-M) bioArchive, Helmholtz Munich, 81377, Munich, Germany
| |
Collapse
|
17
|
Doryab A, Schmid O. Bioactive Cell-Derived ECM Scaffold Forms a Unique Cellular Microenvironment for Lung Tissue Engineering. Biomedicines 2022; 10:biomedicines10081791. [PMID: 35892691 PMCID: PMC9394345 DOI: 10.3390/biomedicines10081791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases are one of the leading causes of death worldwide. Lung transplantation is currently the only causal therapeutic for lung diseases, which is restricted to end-stage disease and limited by low access to donor lungs. Lung tissue engineering (LTE) is a promising approach to regenerating a replacement for at least a part of the damaged lung tissue. Currently, lung regeneration is limited to a simplified local level (e.g., alveolar−capillary barrier) due to the sophisticated and complex structure and physiology of the lung. Here, we introduce an extracellular matrix (ECM)-integrated scaffold using a cellularization−decellularization−recellularization technique. This ECM-integrated scaffold was developed on our artificial co-polymeric BETA (biphasic elastic thin for air−liquid interface cell culture conditions) scaffold, which were initially populated with human lung fibroblasts (IMR90 cell line), as the main generator of ECM proteins. Due to the interconnected porous structure of the thin (<5 µm) BETA scaffold, the cells can grow on and infiltrate into the scaffold and deposit their own ECM. After a mild decellularization procedure, the ECM proteins remained on the scaffold, which now closely mimicked the cellular microenvironment of pulmonary cells more realistically than the plain artificial scaffolds. We assessed several decellularization methods and found that 20 mM NH4OH and 0.1% Triton X100 with subsequent DNase treatment completely removed the fibroblasts (from the first cellularization) and maintains collagen I and IV as the key ECM proteins on the scaffold. We also showed the repopulation of the primary fibroblast from human (without chronic lung disease (non-CLD) donors) and human bronchial epithelial (16HBE14o−) cells on the ECM-integrated BETA scaffold. With this technique, we developed a biomimetic scaffold that can mimic both the physico-mechanical properties and the native microenvironment of the lung ECM. The results indicate the potential of the presented bioactive scaffold for LTE application.
Collapse
|
18
|
Paul A, Kumar S, Kaoud TS, Pickett MR, Bohanon AL, Zoldan J, Dalby KN, Parekh SH. Biomechanical Dependence of SARS-CoV-2 Infections. ACS APPLIED BIO MATERIALS 2022; 5:2307-2315. [PMID: 35486915 PMCID: PMC9063985 DOI: 10.1021/acsabm.2c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022]
Abstract
Older people have been disproportionately vulnerable to the current SARS-CoV-2 pandemic, with an increased risk of severe complications and death compared to other age groups. A mix of underlying factors has been speculated to give rise to this differential infection outcome including changes in lung physiology, weakened immunity, and severe immune response. Our study focuses on the impact of biomechanical changes in lungs that occur as individuals age, that is, the stiffening of the lung parenchyma and increased matrix fiber density. We used hydrogels with an elastic modulus of 0.2 and 50 kPa and conventional tissue culture surfaces to investigate how infection rate changes with parenchymal tissue stiffness in lung epithelial cells challenged with SARS-CoV-2 Spike (S) protein pseudotyped lentiviruses. Further, we employed electrospun fiber matrices to isolate the effect of matrix density. Given the recent data highlighting the importance of alternative virulent strains, we included both the native strain identified in early 2020 and an early S protein variant (D614G) that was shown to increase the viral infectivity markedly. Our results show that cells on softer and sparser scaffolds, closer resembling younger lungs, exhibit higher infection rates by the WT and D614G variant. This suggests that natural changes in lung biomechanics do not increase the propensity for SARS-CoV-2 infection and that other factors, such as a weaker immune system, may contribute to increased disease burden in the elderly.
Collapse
Affiliation(s)
- Alexandra Paul
- Department of Biomedical Engineering,
University of Texas at Austin, Austin, Texas 78712,
United States
- Department of Biology and Biological Engineering,
Chalmers University of Technology, SE-412 98 Gothenburg,
Sweden
| | - Sachin Kumar
- Department of Biomedical Engineering,
University of Texas at Austin, Austin, Texas 78712,
United States
- Centre for Biomedical Engineering, Indian
Institute of Technology Delhi, Hauz Khas, New Delhi 110016,
India
- All India Institute of Medical
Sciences, Ansari Nagar, New Delhi 110029, India
| | - Tamer S. Kaoud
- Division of Chemical Biology and Medicinal Chemistry,
University of Texas at Austin, Austin, Texas 78712,
United States
| | - Madison R. Pickett
- Department of Biomedical Engineering,
University of Texas at Austin, Austin, Texas 78712,
United States
| | - Amanda L. Bohanon
- Division of Chemical Biology and Medicinal Chemistry,
University of Texas at Austin, Austin, Texas 78712,
United States
| | - Janet Zoldan
- Department of Biomedical Engineering,
University of Texas at Austin, Austin, Texas 78712,
United States
| | - Kevin N. Dalby
- Division of Chemical Biology and Medicinal Chemistry,
University of Texas at Austin, Austin, Texas 78712,
United States
| | - Sapun H. Parekh
- Department of Biomedical Engineering,
University of Texas at Austin, Austin, Texas 78712,
United States
| |
Collapse
|
19
|
He Y, Rofaani E, Huang X, Huang B, Liang F, Wang L, Shi J, Peng J, Chen Y. Generation of Alveolar Epithelium Using Reconstituted Basement Membrane and hiPSC-Derived Organoids. Adv Healthc Mater 2022; 11:e2101972. [PMID: 34935309 DOI: 10.1002/adhm.202101972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/01/2021] [Indexed: 12/11/2022]
Abstract
In vitro modeling of alveolar epithelium needs to recapitulate features of both cellular and noncellular components of the lung tissues. Herein, a method is presented to generate alveolar epithelium by using human induced pluripotent stem cells (hiPSCs) and reconstituted or artificial basement membrane (ABM). The ABM is obtained by self-assembling type IV collagen and laminin with a monolayer of crosslinked gelatin nanofibers as backbone and a patterned honeycomb microframe for handling. Alveolar organoids are obtained from hiPSCs and then dissociated into single cells. After replating the alveolar cells on the ABM and a short-period incubation under submerged and air-liquid interface culture conditions, an alveolar epithelium is achieved, showing high-level expressions of both alveolar cell-specific proteins and characteristic tight junctions. Besides, endothelial cells derived from the same hiPSCs are cocultured on the backside of the epithelium, forming an air-blood barrier. The method is generic and can potentially be applied to other types of artificial epithelium and endothelium.
Collapse
Affiliation(s)
- Yong He
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Elrade Rofaani
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Xiaochen Huang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Boxin Huang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Feng Liang
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Li Wang
- MesoBioTech 231 Rue Saint‐Honoré Paris 75001 France
| | - Jian Shi
- MesoBioTech 231 Rue Saint‐Honoré Paris 75001 France
| | - Juan Peng
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| | - Yong Chen
- École Normale Supérieure‐PSL Research University Sorbonne Universités – UPMC Univ Paris 06, CNRS UMR 8640 PASTEUR, 24, rue Lhomond Paris 75005 France
| |
Collapse
|
20
|
Beretta E, Romanò F, Sancini G, Grotberg JB, Nieman GF, Miserocchi G. Pulmonary Interstitial Matrix and Lung Fluid Balance From Normal to the Acutely Injured Lung. Front Physiol 2021; 12:781874. [PMID: 34987415 PMCID: PMC8720972 DOI: 10.3389/fphys.2021.781874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/02/2021] [Indexed: 01/17/2023] Open
Abstract
This review analyses the mechanisms by which lung fluid balance is strictly controlled in the air-blood barrier (ABB). Relatively large trans-endothelial and trans-epithelial Starling pressure gradients result in a minimal flow across the ABB thanks to low microvascular permeability aided by the macromolecular structure of the interstitial matrix. These edema safety factors are lost when the integrity of the interstitial matrix is damaged. The result is that small Starling pressure gradients, acting on a progressively expanding alveolar barrier with high permeability, generate a high transvascular flow that causes alveolar flooding in minutes. We modeled the trans-endothelial and trans-epithelial Starling pressure gradients under control conditions, as well as under increasing alveolar pressure (Palv) conditions of up to 25 cmH2O. We referred to the wet-to-dry weight (W/D) ratio, a specific index of lung water balance, to be correlated with the functional state of the interstitial structure. W/D averages ∼5 in control and might increase by up to ∼9 in severe edema, corresponding to ∼70% loss in the integrity of the native matrix. Factors buffering edemagenic conditions include: (i) an interstitial capacity for fluid accumulation located in the thick portion of ABB, (ii) the increase in interstitial pressure due to water binding by hyaluronan (the "safety factor" opposing the filtration gradient), and (iii) increased lymphatic flow. Inflammatory factors causing lung tissue damage include those of bacterial/viral and those of sterile nature. Production of reactive oxygen species (ROS) during hypoxia or hyperoxia, or excessive parenchymal stress/strain [lung overdistension caused by patient self-induced lung injury (P-SILI)] can all cause excessive inflammation. We discuss the heterogeneity of intrapulmonary distribution of W/D ratios. A W/D ∼6.5 has been identified as being critical for the transition to severe edema formation. Increasing Palv for W/D > 6.5, both trans-endothelial and trans-epithelial gradients favor filtration leading to alveolar flooding. Neither CT scan nor ultrasound can identify this initial level of lung fluid balance perturbation. A suggestion is put forward to identify a non-invasive tool to detect the earliest stages of perturbation of lung fluid balance before the condition becomes life-threatening.
Collapse
Affiliation(s)
- Egidio Beretta
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Francesco Romanò
- Univ. Lille, CNRS, ONERA, Arts et Métiers, Centrale Lille, FRE 2017-LMFL-Laboratoire de Mécanique des Fluides de Lille – Kampé de Fériet, Lille, France
| | - Giulio Sancini
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Gary F. Nieman
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Giuseppe Miserocchi
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| |
Collapse
|
21
|
Mao GC, Gong CC, Wang Z, Sun MX, Pei ZP, Meng WQ, Cen JF, He XW, Lu Y, Xu QQ, Xiao K. BMSC-derived exosomes ameliorate sulfur mustard-induced acute lung injury by regulating the GPRC5A-YAP axis. Acta Pharmacol Sin 2021; 42:2082-2093. [PMID: 33654219 PMCID: PMC8633287 DOI: 10.1038/s41401-021-00625-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sulfur mustard (SM) is a highly toxic chemical warfare agent that causes acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS). There are no effective therapeutic treatments or antidotes available currently to counteract its toxic effects. Our previous study shows that bone marrow-derived mesenchymal stromal cells (BMSCs) could exert therapeutic effects against SM-induced lung injury. In this study, we explored the therapeutic potential of BMSC-derived exosomes (BMSC-Exs) against ALI and the underlying mechanisms. ALI was induced in mice by injection of SM (30 mg/kg, sc) at their medial and dorsal surfaces. BMSC-Exs (20 μg/kg in 200 μL PBS, iv) were injected for a 5-day period after SM exposure. We showed that BMSC-Exs administration caused a protective effect against pulmonary edema. Using a lung epithelial cell barrier model, BMSC-Exs (10, 20, 40 μg) dose-dependently inhibited SM-induced cell apoptosis and promoted the recovery of epithelial barrier function by facilitating the expression and relocalization of junction proteins (E-cadherin, claudin-1, occludin, and ZO-1). We further demonstrated that BMSC-Exs protected against apoptosis and promoted the restoration of barrier function against SM through upregulating G protein-coupled receptor family C group 5 type A (GPRC5A), a retinoic acid target gene predominately expressed in the epithelial cells of the lung. Knockdown of GPRC5A reduced the antiapoptotic and barrier regeneration abilities of BMSC-Exs and diminished their therapeutic effects in vitro and in vivo. BMSC-Exs-caused upregulation of GPRC5A promoted the expression of Bcl-2 and junction proteins via regulating the YAP pathway. In summary, BMSC-Exs treatment exerts protective effects against SM-induced ALI by promoting alveolar epithelial barrier repair and may be an alternative approach to stem cell-based therapy.
Collapse
Affiliation(s)
- Guan-Chao Mao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Chu-Chu Gong
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhen Wang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- Department of Preventive Medicinne, School of Medicine, Hunan Normal University, Changsha, China
| | - Ming-Xue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhi-Peng Pei
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Wen-Qi Meng
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jin-Feng Cen
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xiao-Wen He
- Origincell Technology Group Co., Ltd., Shanghai, 201203, China
| | - Ying Lu
- Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Qing-Qiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Kai Xiao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
22
|
A Robust Protocol for Decellularized Human Lung Bioink Generation Amenable to 2D and 3D Lung Cell Culture. Cells 2021; 10:cells10061538. [PMID: 34207111 PMCID: PMC8234522 DOI: 10.3390/cells10061538] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Decellularization efforts must balance the preservation of the extracellular matrix (ECM) components while eliminating the nucleic acid and cellular components. Following effective removal of nucleic acid and cell components, decellularized ECM (dECM) can be solubilized in an acidic environment with the assistance of various enzymes to develop biological scaffolds in different forms, such as sheets, tubular constructs, or three-dimensional (3D) hydrogels. Each organ or tissue that undergoes decellularization requires a distinct and optimized protocol to ensure that nucleic acids are removed, and the ECM components are preserved. The objective of this study was to optimize the decellularization process for dECM isolation from human lung tissues for downstream 2D and 3D cell culture systems. Following protocol optimization and dECM isolation, we performed experiments with a wide range of dECM concentrations to form human lung dECM hydrogels that were physically stable and biologically responsive. The dECM based-hydrogels supported the growth and proliferation of primary human lung fibroblast cells in 3D cultures. The dECM is also amenable to the coating of polyester membranes in Transwell™ Inserts to improve the cell adhesion, proliferation, and barrier function of primary human bronchial epithelial cells in 2D. In conclusion, we present a robust protocol for human lung decellularization, generation of dECM substrate material, and creation of hydrogels that support primary lung cell viability in 2D and 3D culture systems
Collapse
|
23
|
Jain P, Nishiguchi A, Linz G, Wessling M, Ludwig A, Rossaint R, Möller M, Singh S. Reconstruction of Ultra-thin Alveolar-capillary Basement Membrane Mimics. Adv Biol (Weinh) 2021; 5:e2000427. [PMID: 33987968 DOI: 10.1002/adbi.202000427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/19/2021] [Indexed: 12/15/2022]
Abstract
Alveolar-capillary basement membrane (BM) is ultra-thin (<2 µm) extracellular matrix that maintains integral epithelial-endothelial cell layers. In vitro reconstructions of alveolar-capillary barrier supported on synthetic scaffolds closely resembling the fibrous and ultra-thin natural BM are essential in mimicking the lung pathophysiology. Although BM topology and dimensions are well known to significantly influence cellular behavior, conventionally used BM mimics fail to recreate this natural niche. To overcome this, electrospun ultra-thin 2 µm poly(caprolactone) (PCL) nanofibrous mesh is used to establish an alveolar-capillary barrier model of lung endothelial/epithelial cells. Transepithelial electrical resistance (TEER) and permeability studies reveal integral tight junctions and improved mass transport through the highly porous PCL meshes compared to conventional dense membranes with etched pores. The chemotaxis of neutrophils is shown across the barrier in presence of inflammatory response that is naturally impeded in confined regions. Conventional requirement of 3 µm or larger pore size can lead to barrier disruption due to epithelial/endothelial cell invasion. Despite high porosity, the interconnected BM mimic prevents barrier disruption and allows neutrophil transmigration, thereby demonstrating the physiological relevance of the thin nanofibrous meshes. It is envisioned that these bipolar cultured barriers would contribute to an organ-level in vitro model for pathological disease, environmental pollutants, and nanotoxicology.
Collapse
Affiliation(s)
- Puja Jain
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany
| | - Akihiro Nishiguchi
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany.,Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Georg Linz
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany.,Chemical Process Engineering, RWTH Aachen University, Forckenbeckstr. 51, 52056, Aachen, Germany
| | - Matthias Wessling
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany.,Chemical Process Engineering, RWTH Aachen University, Forckenbeckstr. 51, 52056, Aachen, Germany
| | - Andreas Ludwig
- Institute for Molecular Pharmacology, University Hospital RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, University Hospital RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Martin Möller
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany
| | - Smriti Singh
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52056, Aachen, Germany.,Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| |
Collapse
|
24
|
Munir B, Xu Y. The steady motion of microbubbles in bifurcating airways: Role of shear-thinning and surface tension. Respir Physiol Neurobiol 2021; 290:103675. [PMID: 33915302 DOI: 10.1016/j.resp.2021.103675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/21/2021] [Indexed: 12/11/2022]
Abstract
Mucous fluid is non-Newtonian secretions in the lower lung airways that accumulates when the alveolar-capillary membrane ruptures during acute respiratory distress syndrome. The mucus fluid has, therefore, different types of non-Newtonian properties like shear-thinning, viscoelasticity, and non-zero yield stress. In this paper, we numerically solved the steady Stokes equations along with arbitrary Eulerian-Lagrangian moving mesh techniques to study the microbubble propagation in a two-dimensional asymmetric bifurcating airway filled with non-Newtonian fluid where the fluid has shear-thinning behavior described by the power-law model. Numerical results show that both shear-thinning and surface tension characterized by the behavior index (n) and Capillary number (Ca), respectively, had a significant impact on microbubble flow patterns and the magnitude of the pressure gradient. At low values of both n and Ca, the microbubble leaves a thin film-thickness with the airway wall while a large and sharp peak of the pressure gradient near the thin bubble tip. Interestingly, increasing both n and Ca, leads to an increase in film thickness and a decrease in the pressure gradient magnitude in both the daughter airway walls. It is observed the magnitude of the pressure gradient is more sensitive to Ca compared to n. We concluded that shear-thinning and surface tension not only significantly impact the patterns of microbubble propagation but also the hydrodynamic stress magnitudes at the airway wall.
Collapse
Affiliation(s)
- Bacha Munir
- School of Natural and Applied Sciences, Department of Applied Mathematics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710029, People's Republic of China.
| | - Yong Xu
- School of Natural and Applied Sciences, Department of Applied Mathematics, Northwestern Polytechnical University, Xi'an, Shaanxi, 710029, People's Republic of China
| |
Collapse
|
25
|
Chen G, Ge D, Zhu B, Shi H, Ma Q. Upregulation of matrix metalloproteinase 9 (MMP9)/tissue inhibitor of metalloproteinase 1 (TIMP1) and MMP2/TIMP2 ratios may be involved in lipopolysaccharide-induced acute lung injury. J Int Med Res 2021; 48:300060520919592. [PMID: 32339071 PMCID: PMC7219017 DOI: 10.1177/0300060520919592] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective This study aimed to examine the changes and significance of matrix metalloproteinase 9 (MMP9), MMP2, tissue inhibitor of metalloproteinase 1 (TIMP1), and TIMP2 in rats with lipopolysaccharide (LPS)-induced acute lung injury (ALI). Methods Wistar rats were randomly divided into a control group (injected with saline) and an ALI group (injected with LPS), then subdivided into four time points (2, 6, 12, and 24 hours). Serum tumor necrosis factor alpha and interleukin-6 levels were detected by ELISA to investigate the inflammatory reaction after LPS injection. The degree of ALI was determined by hematoxylin–eosin staining of lung tissue, the lung wet/dry weight ratio, and pulmonary permeability index. Changes in lung MMP and TIMP protein and mRNA levels were detected by western blotting and quantitative real-time polymerase chain reaction. Results Changes in the ratios of MMP9/TIMP1 and MMP2/TIMP2 were consistent with and strongly positively associated with the lung wet/dry weight ratio, the pulmonary permeability index, and serum tumor necrosis factor alpha and interleukin-6 levels in the ALI group. Conclusion ALI induced by LPS may be related to upregulation of MMP9/TIMP1 and MMP2/TIMP2 ratios.
Collapse
Affiliation(s)
- Guobing Chen
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Dandan Ge
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Pediatric Key Laboratory of Xiamen, Xiamen, Fujian, China
| | - Bizhen Zhu
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Huixuan Shi
- Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Qilin Ma
- School of Medicine, Xiamen University, Xiamen, Fujian, China.,Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
26
|
Protective Effects of Kirenol against Lipopolysaccharide-Induced Acute Lung Injury through the Modulation of the Proinflammatory NFκB Pathway and the AMPK2-/Nrf2-Mediated HO-1/AOE Pathway. Antioxidants (Basel) 2021; 10:antiox10020204. [PMID: 33572510 PMCID: PMC7911485 DOI: 10.3390/antiox10020204] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury (ALI) is an acute and life-threatening inflammatory disease of the lung parenchyma that is associated with high mortality worldwide. No therapeutic strategies have been developed for the mitigation of the proinflammatory response that characterizes ALI. Kirenol has anti-inflammatory, antiarthritic, and immunoregulatory effects. In the present study, we investigated the protective effects of kirenol against lipopolysaccharides (LPS)-induced ALI in mice. Kirenol reduced the LPS-induced histopathology changes involving edema and thickening of the interstitial or alveolar walls, infiltration of leukocytes, formation of hyaline membrane. Pretreatment with kirenol reduced leukocytes infiltration in bronchoalveolar lavage fluid (BALF), the alveolar-capillary barrier disruption and lipid peroxidation in lung tissues induced by LPS. Kirenol significantly inhibited the secretion of cytokines, IL-1β, IL6, and TNFα, into the BALF of the mice with LPS-induced ALI through NFκB activation. Moreover, kirenol attenuated the downregulation of the antioxidant enzymes, superoxide dismutase, glutathione peroxidase, and catalase that was induced by LPS. HO-1 expression and the phosphorylation of Nrf2 and AMPK2 were also induced by kirenol. The results indicate that kirenol can be developed as a treatment strategy for ALI, and its effects are induced through the inhibition of the NF-κB proinflammatory pathway and promotion of AMPK2/Nrf2-mediated HO-1 and antioxidant enzymes (AOE) activation.
Collapse
|
27
|
Doryab A, Taskin MB, Stahlhut P, Schröppel A, Orak S, Voss C, Ahluwalia A, Rehberg M, Hilgendorff A, Stöger T, Groll J, Schmid O. A Bioinspired in vitro Lung Model to Study Particokinetics of Nano-/Microparticles Under Cyclic Stretch and Air-Liquid Interface Conditions. Front Bioeng Biotechnol 2021; 9:616830. [PMID: 33634087 PMCID: PMC7902031 DOI: 10.3389/fbioe.2021.616830] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Evolution has endowed the lung with exceptional design providing a large surface area for gas exchange area (ca. 100 m2) in a relatively small tissue volume (ca. 6 L). This is possible due to a complex tissue architecture that has resulted in one of the most challenging organs to be recreated in the lab. The need for realistic and robust in vitro lung models becomes even more evident as causal therapies, especially for chronic respiratory diseases, are lacking. Here, we describe the Cyclic InVItroCell-stretch (CIVIC) “breathing” lung bioreactor for pulmonary epithelial cells at the air-liquid interface (ALI) experiencing cyclic stretch while monitoring stretch-related parameters (amplitude, frequency, and membrane elastic modulus) under real-time conditions. The previously described biomimetic copolymeric BETA membrane (5 μm thick, bioactive, porous, and elastic) was attempted to be improved for even more biomimetic permeability, elasticity (elastic modulus and stretchability), and bioactivity by changing its chemical composition. This biphasic membrane supports both the initial formation of a tight monolayer of pulmonary epithelial cells (A549 and 16HBE14o−) under submerged conditions and the subsequent cell-stretch experiments at the ALI without preconditioning of the membrane. The newly manufactured versions of the BETA membrane did not improve the characteristics of the previously determined optimum BETA membrane (9.35% PCL and 6.34% gelatin [w/v solvent]). Hence, the optimum BETA membrane was used to investigate quantitatively the role of physiologic cyclic mechanical stretch (10% linear stretch; 0.33 Hz: light exercise conditions) on size-dependent cellular uptake and transepithelial transport of nanoparticles (100 nm) and microparticles (1,000 nm) for alveolar epithelial cells (A549) under ALI conditions. Our results show that physiologic stretch enhances cellular uptake of 100 nm nanoparticles across the epithelial cell barrier, but the barrier becomes permeable for both nano- and micron-sized particles (100 and 1,000 nm). This suggests that currently used static in vitro assays may underestimate cellular uptake and transbarrier transport of nanoparticles in the lung.
Collapse
Affiliation(s)
- Ali Doryab
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Mehmet Berat Taskin
- Department of Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Philipp Stahlhut
- Department of Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Andreas Schröppel
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Sezer Orak
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Carola Voss
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Arti Ahluwalia
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Markus Rehberg
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany.,Center for Comprehensive Developmental Care (CDeCLMU), Dr. von Haunersches Children's Hospital University, Hospital of the Ludwig-Maximilians University, Munich, Germany
| | - Tobias Stöger
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Otmar Schmid
- Comprehensive Pneumology Center Munich, Member of the German Center for Lung Research, Munich, Germany.,Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Lung Biology and Disease, Munich, Germany
| |
Collapse
|
28
|
Bobba CM, Fei Q, Shukla V, Lee H, Patel P, Putman RK, Spitzer C, Tsai M, Wewers MD, Lee RJ, Christman JW, Ballinger MN, Ghadiali SN, Englert JA. Nanoparticle delivery of microRNA-146a regulates mechanotransduction in lung macrophages and mitigates injury during mechanical ventilation. Nat Commun 2021; 12:289. [PMID: 33436554 PMCID: PMC7804938 DOI: 10.1038/s41467-020-20449-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/03/2020] [Indexed: 12/15/2022] Open
Abstract
Mechanical ventilation generates injurious forces that exacerbate lung injury. These forces disrupt lung barrier integrity, trigger proinflammatory mediator release, and differentially regulate genes and non-coding oligonucleotides including microRNAs. In this study, we identify miR-146a as a mechanosensitive microRNA in alveolar macrophages that has therapeutic potential to mitigate lung injury during mechanical ventilation. We use humanized in-vitro systems, mouse models, and biospecimens from patients to elucidate the expression dynamics of miR-146a needed to decrease lung injury during mechanical ventilation. We find that the endogenous increase in miR-146a following injurious ventilation is not sufficient to prevent lung injury. However, when miR-146a is highly overexpressed using a nanoparticle delivery platform it is sufficient to prevent injury. These data indicate that the endogenous increase in microRNA-146a during mechanical ventilation is a compensatory response that partially limits injury and that nanoparticle delivery of miR-146a is an effective strategy for mitigating lung injury during mechanical ventilation.
Collapse
Affiliation(s)
- Christopher M Bobba
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Qinqin Fei
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH, 43210, USA
| | - Vasudha Shukla
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Hyunwook Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Pragi Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Rachel K Putman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Carleen Spitzer
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - MuChun Tsai
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Mark D Wewers
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, OH, 43210, USA
| | - John W Christman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Megan N Ballinger
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA
| | - Samir N Ghadiali
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA.
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
- Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue, Columbus, OH, 43210, USA.
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, 473 West 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
29
|
Salvia miltiorrhiza Injection Alleviates LPS-Induced Acute Lung Injury by Adjusting the Balance of MMPs/TIMPs Ratio. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9617081. [PMID: 32765635 PMCID: PMC7387992 DOI: 10.1155/2020/9617081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/10/2020] [Accepted: 07/01/2020] [Indexed: 11/26/2022]
Abstract
Salvia miltiorrhiza injection (SMI) is a classical traditional Chinese medicine, which plays an active role in the treatment of many diseases such as promoting blood circulation, removing blood stasis, reducing inflammatory reaction, and improving acute lung injury (ALI). Previous studies have shown that matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) are involved in the pathophysiological process of ALI. However, the relationship between SMI and MMPs/TIMPs remains unclear. In this study, Wistar rats were randomly divided into control group (NC), Salvia miltiorrhiza group (SM), lipopolysaccharide group (LPS), and Salvia miltiorrhiza treatment group (Tsm). The four groups were subdivided into four time points (2, 6, 12, and 24 hours), and specimens were collected after animal sacrifice at each time point. Serum TNF-α and IL-6 levels were detected by ELISA. The degree of lung injury was determined by lung tissue hematoxylin-eosin staining, lung wet/dry weight (W/D) ratio, and lung permeability index. The changes in lung MMPs/TIMPs protein and mRNA were detected by Western blot and real-time quantitative PCR. The results showed that rats injected with LPS experience acute lung injury, and the ratio of MMPs/TIMPs in lung tissues increased gradually with time. In the Tsm group, the ratio of MMPs/TIMPs decreased gradually, and likewise, the balance was gradually restored, while indicators related to lung injury were gradually declined. These data suggest that SMI alleviates LPS-induced acute lung injury; this protective effect may be related to regulation of the balance of MMPs/TIMPs ratio.
Collapse
|
30
|
Munir B, Xu Y. Effects of gravity and surface tension on steady microbubble propagation in asymmetric bifurcating airways. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2020; 32:072105. [PMID: 35002196 PMCID: PMC8722330 DOI: 10.1063/5.0012796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/03/2020] [Indexed: 05/21/2023]
Abstract
Mechanical ventilation is nowadays a well-developed, safe, and necessary strategy for acute respiratory distress syndrome patients to survive. However, the propagation of microbubbles in airway bifurcations during mechanical ventilation makes the existing lung injury more severe. In this paper, finite element and direct interface tracking techniques were utilized to simulate steady microbubble propagation in a two-dimensional asymmetric bifurcating airway filled with a viscous fluid. Inertial effects were neglected, and the numerical solution of Stokes's equations was used to investigate how gravity and surface tension defined by a Bond (Bo) number and capillary (Ca) number influence the magnitudes of pressure gradients, shear stresses, and shear stress gradients on the bifurcating daughter airway wall. It is found that increasing Bo significantly influenced both the bubble shape and hydrodynamic stresses, where Bo ≥ 0.25 results in a significant increase in bubble elevation and pressure gradient in the upper daughter wall. Although for both Bo and Ca, the magnitude of the pressure gradient is always much larger in the upper daughter airway wall, Ca has a great role in amplifying the magnitude of the pressure gradient. In conclusion, both gravity and surface tension play a key role in the steady microbubble propagation and hydrodynamic stresses in the bifurcating airways.
Collapse
Affiliation(s)
- Bacha Munir
- Author to whom correspondence should be addressed:
| | | |
Collapse
|
31
|
Radiom M, He Y, Peng-Wang J, Baeza-Squiban A, Berret JF, Chen Y. Alveolar mimics with periodic strain and its effect on the cell layer formation. Biotechnol Bioeng 2020; 117:2827-2841. [PMID: 32542664 DOI: 10.1002/bit.27458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/09/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
We report on the development of a new model of alveolar air-tissue interface on a chip. The model consists of an array of suspended hexagonal monolayers of gelatin nanofibers supported by microframes and a microfluidic device for the patch integration. The suspended monolayers are deformed to a central displacement of 40-80 µm at the air-liquid interface by application of air pressure in the range of 200-1,000 Pa. With respect to the diameter of the monolayers, that is, 500 µm, this displacement corresponds to a linear strain of 2-10% in agreement with the physiological strain range in the lung alveoli. The culture of A549 cells on the monolayers for an incubation time of 1-3 days showed viability in the model. We exerted a periodic strain of 5% at a frequency of 0.2 Hz for 1 hr to the cells. We found that the cells were strongly coupled to the nanofibers, but the strain reduced the coupling and induced remodeling of the actin cytoskeleton, which led to a better tissue formation. Our model can serve as a versatile tool in lung investigations such as in inhalation toxicology and therapy.
Collapse
Affiliation(s)
- Milad Radiom
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057, Université Paris Diderot Paris-VII, Paris, France
| | - Yong He
- Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, Pasteur, École Normale Supérieure-PSL Research University, Paris, France
| | - Juan Peng-Wang
- Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, Pasteur, École Normale Supérieure-PSL Research University, Paris, France
| | - Armelle Baeza-Squiban
- Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Université Paris Diderot Paris-VII, Paris, France
| | - Jean-François Berret
- Laboratoire Matière et Systèmes Complexes, CNRS UMR 7057, Université Paris Diderot Paris-VII, Paris, France
| | - Yong Chen
- Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, Pasteur, École Normale Supérieure-PSL Research University, Paris, France
| |
Collapse
|
32
|
Abalymov A, Parakhonskiy B, Skirtach AG. Polymer- and Hybrid-Based Biomaterials for Interstitial, Connective, Vascular, Nerve, Visceral and Musculoskeletal Tissue Engineering. Polymers (Basel) 2020; 12:E620. [PMID: 32182751 PMCID: PMC7182904 DOI: 10.3390/polym12030620] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review, materials based on polymers and hybrids possessing both organic and inorganic contents for repairing or facilitating cell growth in tissue engineering are discussed. Pure polymer based biomaterials are predominantly used to target soft tissues. Stipulated by possibilities of tuning the composition and concentration of their inorganic content, hybrid materials allow to mimic properties of various types of harder tissues. That leads to the concept of "one-matches-all" referring to materials possessing the same polymeric base, but different inorganic content to enable tissue growth and repair, proliferation of cells, and the formation of the ECM (extra cellular matrix). Furthermore, adding drug delivery carriers to coatings and scaffolds designed with such materials brings additional functionality by encapsulating active molecules, antibacterial agents, and growth factors. We discuss here materials and methods of their assembly from a general perspective together with their applications in various tissue engineering sub-areas: interstitial, connective, vascular, nervous, visceral and musculoskeletal tissues. The overall aims of this review are two-fold: (a) to describe the needs and opportunities in the field of bio-medicine, which should be useful for material scientists, and (b) to present capabilities and resources available in the area of materials, which should be of interest for biologists and medical doctors.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | | | - Andre G. Skirtach
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
33
|
Viola H, Chang J, Grunwell JR, Hecker L, Tirouvanziam R, Grotberg JB, Takayama S. Microphysiological systems modeling acute respiratory distress syndrome that capture mechanical force-induced injury-inflammation-repair. APL Bioeng 2019; 3:041503. [PMID: 31768486 PMCID: PMC6874511 DOI: 10.1063/1.5111549] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Complex in vitro models of the tissue microenvironment, termed microphysiological systems, have enormous potential to transform the process of discovering drugs and disease mechanisms. Such a paradigm shift is urgently needed in acute respiratory distress syndrome (ARDS), an acute lung condition with no successful therapies and a 40% mortality rate. Here, we consider how microphysiological systems could improve understanding of biological mechanisms driving ARDS and ultimately improve the success of therapies in clinical trials. We first discuss how microphysiological systems could explain the biological mechanisms underlying the segregation of ARDS patients into two clinically distinct phenotypes. Then, we contend that ARDS-mimetic microphysiological systems should recapitulate three critical aspects of the distal airway microenvironment, namely, mechanical force, inflammation, and fibrosis, and we review models that incorporate each of these aspects. Finally, we recognize the substantial challenges associated with combining inflammation, fibrosis, and/or mechanical force in microphysiological systems. Nevertheless, complex in vitro models are a novel paradigm for studying ARDS, and they could ultimately improve patient care.
Collapse
Affiliation(s)
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA
| | - Jocelyn R. Grunwell
- Department of Pediatrics, Division of Critical Care Medicine, Children's Healthcare of Atlanta at Egleston, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona, Tucson, Arizona 85724, USA and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona 85723, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA and Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia 30322, USA
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
34
|
Young BM, Shankar K, Tho CK, Pellegrino AR, Heise RL. Laminin-driven Epac/Rap1 regulation of epithelial barriers on decellularized matrix. Acta Biomater 2019; 100:223-234. [PMID: 31593773 DOI: 10.1016/j.actbio.2019.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 12/28/2022]
Abstract
Decellularized tissues offer a unique tool for developing regenerative biomaterials or in vitro platforms for the study of cell-extracellular matrix (ECM) interactions. One main challenge associated with decellularized lung tissue is that ECM components can be stripped away or altered by the detergents used to remove cellular debris. Without characterizing the composition of lung decellularized ECM (dECM) and the cellular response caused by the altered composition, it is difficult to utilize dECM for regeneration and specifically, engineering the complexities of the alveolar-capillary barrier. This study takes steps towards uncovering if dECM must be enhanced with lost ECM proteins to achieve proper epithelial barrier formation. To achieve this, the epithelial barrier function was assessed on dECM coatings with and without the systematic addition of several key basement membrane proteins. After comparing barrier function on collagen I, fibronectin, laminin, and dECM in varying combinations as an in vitro coating, the alveolar epithelium exhibited superior barrier function when dECM was supplemented with laminin as evidenced by trans-epithelial electrical resistance (TEER) and permeability assays. Increased barrier resistance with laminin addition was associated with upregulation of Claudin-18, E-cadherin, and junction adhesion molecule (JAM)-A, and stabilization of zonula occludens (ZO)-1 at junction complexes. The Epac/Rap1 pathway was observed to play a role in the ECM-mediated barrier function determined by protein expression and Epac inhibition. These findings revealed potential ECM coatings and molecular therapeutic targets for improved regeneration with decellularized scaffolds. STATEMENT OF SIGNIFICANCE: Efforts to produce a transplantable organ-scale biomaterial for lung regeneration has not been entirely successful to date, due to incomplete cell-cell junction formation, ultimately leading to severe edema in vivo. To fully understand the process of alveolar junction formation on ECM-derived biomaterials, this research has characterized and tailored decellularized ECM (dECM) to mitigate reductions in barrier strength or cell attachment caused by abnormal ECM compositions or detergent damage to dECM. These results indicate that laminin-driven Epac signaling plays a vital role in the stabilization of the alveolar barrier. Addition of laminin or Epac agonists during alveolar regeneration can reduce epithelial permeability within bioengineered lungs.
Collapse
Affiliation(s)
- Bethany M Young
- Department of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh St, Room 1071, Richmond, VA 23219, United States
| | - Keerthana Shankar
- Department of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh St, Room 1071, Richmond, VA 23219, United States
| | - Cindy K Tho
- Department of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh St, Room 1071, Richmond, VA 23219, United States
| | - Amanda R Pellegrino
- Department of Biomedical Engineering and Nursing, Duquesne University, 600 Forbes Ave, Pittsburg, Pennsylvania 15282, United States
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh St, Room 1071, Richmond, VA 23219, United States; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall St, Richmond, Virginia 23298, United States.
| |
Collapse
|
35
|
Permeability of Epithelial/Endothelial Barriers in Transwells and Microfluidic Bilayer Devices. MICROMACHINES 2019; 10:mi10080533. [PMID: 31412604 PMCID: PMC6722679 DOI: 10.3390/mi10080533] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/08/2019] [Accepted: 08/11/2019] [Indexed: 12/13/2022]
Abstract
Lung-on-a-chip (LoC) models hold the potential to rapidly change the landscape for pulmonary drug screening and therapy, giving patients more advanced and less invasive treatment options. Understanding the drug absorption in these microphysiological systems, modeling the lung-blood barrier is essential for increasing the role of the organ-on-a-chip technology in drug development. In this work, epithelial/endothelial barrier tissue interfaces were established in microfluidic bilayer devices and transwells, with porous membranes, for permeability characterization. The effect of shear stress on the molecular transport was assessed using known paracellular and transcellular biomarkers. The permeability of porous membranes without cells, in both models, is inversely proportional to the molecular size due to its diffusivity. Paracellular transport, between epithelial/endothelial cell junctions, of large molecules such as transferrin, as well as transcellular transport, through cell lacking required active transporters, of molecules such as dextrans, is negligible. When subjected to shear stress, paracellular transport of intermediate-size molecules such as dextran was enhanced in microfluidic devices when compared to transwells. Similarly, shear stress enhances paracellular transport of small molecules such as Lucifer yellow, but its effect on transcellular transport is not clear. The results highlight the important role that LoC can play in drug absorption studies to accelerate pulmonary drug development.
Collapse
|
36
|
Sedláková V, Kloučková M, Garlíková Z, Vašíčková K, Jaroš J, Kandra M, Kotasová H, Hampl A. Options for modeling the respiratory system: inserts, scaffolds and microfluidic chips. Drug Discov Today 2019; 24:971-982. [PMID: 30877077 DOI: 10.1016/j.drudis.2019.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/08/2019] [Accepted: 03/06/2019] [Indexed: 12/29/2022]
Abstract
The human respiratory system is continuously exposed to varying levels of hazardous substances ranging from environmental toxins to purposely administered drugs. If the noxious effects exceed the inherent regenerative capacity of the respiratory system, injured tissue undergoes complex remodeling that can significantly affect lung function and lead to various diseases. Advanced near-to-native in vitro lung models are required to understand the mechanisms involved in pulmonary damage and repair and to reliably test the toxicity of compounds to lung tissue. This review is an overview of the development of in vitro respiratory system models used for study of lung diseases. It includes discussion of using these models for environmental toxin assessment and pulmonary toxicity screening.
Collapse
Affiliation(s)
- Veronika Sedláková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; Division of Cardiac Surgery, Cardiovascular Tissue Engineering Laboratory, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa K1Y 4W7, Canada.
| | - Michaela Kloučková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - Zuzana Garlíková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; International Clinical Research Center, St Anne's University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Kateřina Vašíčková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; International Clinical Research Center, St Anne's University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Josef Jaroš
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; International Clinical Research Center, St Anne's University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Mário Kandra
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; International Clinical Research Center, St Anne's University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Hana Kotasová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; International Clinical Research Center, St Anne's University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| |
Collapse
|
37
|
Montesanto S, Smithers NP, Bucchieri F, Brucato V, La Carrubba V, Davies DE, Conforti F. Establishment of a pulmonary epithelial barrier on biodegradable poly-L-lactic-acid membranes. PLoS One 2019; 14:e0210830. [PMID: 30653572 PMCID: PMC6336298 DOI: 10.1371/journal.pone.0210830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/20/2018] [Indexed: 11/18/2022] Open
Abstract
Development of biocompatible and functional scaffolds for tissue engineering is a major challenge, especially for development of polarised epithelia that are critical structures in tissue homeostasis. Different in vitro models of the lung epithelial barrier have been characterized using non-degradable polyethylene terephthalate membranes which limits their uses for tissue engineering. Although poly-L-lactic acid (PLLA) membranes are biodegradable, those prepared via conventional Diffusion Induced Phase Separation (DIPS) lack open-porous geometry and show limited permeability compromising their use for epithelial barrier studies. Here we used PLLA membranes prepared via a modification of the standard DIPS protocol to control the membrane surface morphology and permeability. These were bonded to cell culture inserts for use in barrier function studies. Pulmonary epithelial cells (H441) readily attached to the PLLA membranes and formed a confluent cell layer within two days. This was accompanied by a significant increase in trans-epithelial electrical resistance and correlated with the formation of tight junctions and vectorial cytokine secretion in response to TNFα. Our data suggest that a structurally polarized and functional epithelial barrier can be established on PLLA membranes produced via a non-standard DIPS protocol. Therefore, PLLA membranes have potential utility in lung tissue engineering applications requiring bio-absorbable membranes.
Collapse
Affiliation(s)
- Salvatore Montesanto
- The Brooke Laboratory, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Department of Civil, Environmental, Aerospace, Materials Engineering (DICAM), University of Palermo, Palermo, Italy
| | - Natalie P. Smithers
- The Brooke Laboratory, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo, Palermo, Italy
| | - Valerio Brucato
- Department of Civil, Environmental, Aerospace, Materials Engineering (DICAM), University of Palermo, Palermo, Italy
- Interuniversitary Consortium for Material Science and Technology (INSTM) – Research Unit of Palermo, Palermo, Italy
| | - Vincenzo La Carrubba
- Department of Civil, Environmental, Aerospace, Materials Engineering (DICAM), University of Palermo, Palermo, Italy
- Interuniversitary Consortium for Material Science and Technology (INSTM) – Research Unit of Palermo, Palermo, Italy
| | - Donna E. Davies
- The Brooke Laboratory, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, United Kingdom
| | - Franco Conforti
- The Brooke Laboratory, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|