1
|
Torkashvand M, Rezakhani L, Habibi Z, Mikaeili A, Rahmati S. Innovative approaches in lung tissue engineering: the role of exosome-loaded bioscaffolds in regenerative medicine. Front Bioeng Biotechnol 2024; 12:1502155. [PMID: 39758953 PMCID: PMC11695380 DOI: 10.3389/fbioe.2024.1502155] [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: 09/26/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Lung diseases account for over four million premature deaths every year, and experts predict that this number will increase in the future. The top cause of death globally is diseases which include conditions like lung cancer asthma and COPD. Treating severe acute lung injury is a complex task because lungs struggle to heal themselves in the presence of swelling inflammation and scarring caused by damage, to the lung tissues. Though achieving lung regeneration, in controlled environments is still an ambition; ongoing studies are concentrating on notable progress, in the field of lung tissue engineering and methods for repairing lung damage. This review delves into methods, for regenerating lungs with a focus on exosome carry bioscaffolds and mesenchymal stem cells among others. It talks about how these new techniques can help repair lung tissue and improve lung function in cases of damage. Also noted is the significance of ex vivo lung perfusion (EVLP), for rejuvenating donor lungs and the healing properties of exosomes in supporting lung regeneration.
Collapse
Affiliation(s)
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Habibi
- Clinical Research Development Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Abdolhamid Mikaeili
- Medical Biology Research Center, Health Technology Institute, University of Medical Sciences, Kermanshah, Iran
| | - Shima Rahmati
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
2
|
Malmros K, Kirova N, Kotarsky H, Carlsén D, Mansour MSI, Magnusson M, Prabhala P, Brunnström H. 3D cultivation of non-small-cell lung cancer cell lines using four different methods. J Cancer Res Clin Oncol 2024; 150:472. [PMID: 39441367 PMCID: PMC11499447 DOI: 10.1007/s00432-024-06003-x] [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: 08/22/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE The aim of this study was to set up reliable and reproducible culture conditions for 3D tumoroids derived from non-small cell lung cancer (NSCLC) cell lines to enable greater opportunity for successful cultivation of patient-derived samples. METHODS Four NSCLC cell lines, two adenocarcinomas (A549, NCI-H1975) and two squamous cell carcinomas (HCC-95, HCC-1588), were first cultured in traditional 2D settings. Their expected expression profiles concerning TTF-1, CK7, CK5, and p40 status were confirmed by immunohistochemistry (IHC) before the generation of 3D cultures. Tumoroids were established in the hydrogel GrowDex®-T, Nunclon™ Sphera™ flasks, BIOFLOAT™ plates, and Corning® Elplasia® plates. Western blot was used to verify antigen protein expression. Hematoxylin-eosin staining was used to evaluate the cell morphology in the 2D and 3D cultures. Mutational analysis of KRAS and EGFR by PCR on extracted DNA from 3D tumoroids generated from cells with known mutations (A549; KRAS G12S mutation, NCI-H1975; EGFR L858R/T790M mutations). RESULTS We successfully established 3D cultures from A549, NCI-H1975, HCC-95, and HCC-1588 with all four used cultivation methods. The adenocarcinomas (A549, NCI-H1975) maintained their original IHC features in the tumoroids, while the squamous cell carcinomas (HCC-95, HCC-1588) lost their unique markers in the cultures. PCR analysis confirmed persistent genetic changes where expected. CONCLUSION The establishment of tumoroids from lung cancer cell lines is feasible with various methodologies, which is promising for future tumoroid growth from clinical lung cancer samples. However, analysis of relevant markers is a prerequisite and may need to be validated for each model and cell type.
Collapse
Affiliation(s)
- Karina Malmros
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, BMC B13, Klinikgatan 26, Lund, SE-221 00, Sweden.
| | - Nadi Kirova
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, BMC B13, Klinikgatan 26, Lund, SE-221 00, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Skåne University Hospital Lund, Lund, SE-221 85, Sweden
| | - Heike Kotarsky
- Department of Genetics, Pathology, and Molecular Diagnostics, Skåne University Hospital Lund, Lund, SE-221 85, Sweden
| | - Daniel Carlsén
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, BMC B13, Klinikgatan 26, Lund, SE-221 00, Sweden
| | - Mohammed S I Mansour
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, BMC B13, Klinikgatan 26, Lund, SE-221 00, Sweden
- Department of Pathology and Cytology, Halland Hospital Halmstad, Halmstad, SE-301 85, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, SE-221 00, Sweden
| | - Pavan Prabhala
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, SE-221 00, Sweden
| | - Hans Brunnström
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, BMC B13, Klinikgatan 26, Lund, SE-221 00, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Skåne University Hospital Lund, Lund, SE-221 85, Sweden
| |
Collapse
|
3
|
Loncarevic I, Mutlu S, Dzepic M, Keshavan S, Petri-Fink A, Blank F, Rothen-Rutishauser B. Current Challenges to Align Inflammatory Key Events in Animals and Lung Cell Models In Vitro. Chem Res Toxicol 2024; 37:1601-1611. [PMID: 39115970 PMCID: PMC11497357 DOI: 10.1021/acs.chemrestox.4c00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
With numerous novel and innovative in vitro models emerging every year to reduce or replace animal testing, there is an urgent need to align the design, harmonization, and validation of such systems using in vitro-in vivo extrapolation (IVIVE) approaches. In particular, in inhalation toxicology, there is a lack of predictive and prevalidated in vitro lung models that can be considered a valid alternative for animal testing. The predictive power of such models can be enhanced by applying the Adverse Outcome Pathways (AOP) framework, which casually links key events (KE) relevant to IVIVE. However, one of the difficulties identified is that the endpoint analysis and readouts of specific assays in in vitro and animal models for specific toxicants are currently not harmonized, making the alignment challenging. We summarize the current state of the art in endpoint analysis in the two systems, focusing on inflammatory-induced effects and providing guidance for future research directions to improve the alignment.
Collapse
Affiliation(s)
- Isidora Loncarevic
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Seyran Mutlu
- Lung
Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department
for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University
of Bern, Bern, Switzerland
| | - Martina Dzepic
- Lung
Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department
for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University
of Bern, Bern, Switzerland
| | - Sandeep Keshavan
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe
Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Chemistry
Department, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Fabian Blank
- Lung
Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department
for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University
of Bern, Bern, Switzerland
| | | |
Collapse
|
4
|
Mahieu L, Van Moll L, De Vooght L, Delputte P, Cos P. In vitro modelling of bacterial pneumonia: a comparative analysis of widely applied complex cell culture models. FEMS Microbiol Rev 2024; 48:fuae007. [PMID: 38409952 PMCID: PMC10913945 DOI: 10.1093/femsre/fuae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/29/2024] [Accepted: 02/24/2024] [Indexed: 02/28/2024] Open
Abstract
Bacterial pneumonia greatly contributes to the disease burden and mortality of lower respiratory tract infections among all age groups and risk profiles. Therefore, laboratory modelling of bacterial pneumonia remains important for elucidating the complex host-pathogen interactions and to determine drug efficacy and toxicity. In vitro cell culture enables for the creation of high-throughput, specific disease models in a tightly controlled environment. Advanced human cell culture models specifically, can bridge the research gap between the classical two-dimensional cell models and animal models. This review provides an overview of the current status of the development of complex cellular in vitro models to study bacterial pneumonia infections, with a focus on air-liquid interface models, spheroid, organoid, and lung-on-a-chip models. For the wide scale, comparative literature search, we selected six clinically highly relevant bacteria (Pseudomonas aeruginosa, Mycoplasma pneumoniae, Haemophilus influenzae, Mycobacterium tuberculosis, Streptococcus pneumoniae, and Staphylococcus aureus). We reviewed the cell lines that are commonly used, as well as trends and discrepancies in the methodology, ranging from cell infection parameters to assay read-outs. We also highlighted the importance of model validation and data transparency in guiding the research field towards more complex infection models.
Collapse
Affiliation(s)
- Laure Mahieu
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Laurence Van Moll
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Linda De Vooght
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Paul Cos
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
5
|
Kurow O, Nuwayhid R, Stock P, Steinert M, Langer S, Krämer S, Metelmann IB. Organotypic 3D Co-Culture of Human Pleura as a Novel In Vitro Model of Staphylococcus aureus Infection and Biofilm Development. Bioengineering (Basel) 2023; 10:bioengineering10050537. [PMID: 37237611 DOI: 10.3390/bioengineering10050537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Bacterial pleural infections are associated with high mortality. Treatment is complicated due to biofilm formation. A common causative pathogen is Staphylococcus aureus (S. aureus). Since it is distinctly human-specific, rodent models do not provide adequate conditions for research. The purpose of this study was to examine the effects of S. aureus infection on human pleural mesothelial cells using a recently established 3D organotypic co-culture model of pleura derived from human specimens. After infection of our model with S. aureus, samples were harvested at defined time points. Histological analysis and immunostaining for tight junction proteins (c-Jun, VE-cadherin, and ZO-1) were performed, demonstrating changes comparable to in vivo empyema. The measurement of secreted cytokine levels (TNF-α, MCP-1, and IL-1β) proved host-pathogen interactions in our model. Similarly, mesothelial cells produced VEGF on in vivo levels. These findings were contrasted by vital, unimpaired cells in a sterile control model. We were able to establish a 3D organotypic in vitro co-culture model of human pleura infected with S. aureus resulting in the formation of biofilm, including host-pathogen interactions. This novel model could be a useful microenvironment tool for in vitro studies on biofilm in pleural empyema.
Collapse
Affiliation(s)
- Olga Kurow
- Department of Orthopedic, Trauma and Plastic Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Rima Nuwayhid
- Department of Orthopedic, Trauma and Plastic Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Peggy Stock
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Matthias Steinert
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Stefan Langer
- Department of Orthopedic, Trauma and Plastic Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Sebastian Krämer
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| | - Isabella B Metelmann
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
6
|
Abstract
Tumor metastasis is a multiple cascade process where tumor cells disseminate from the primary site to distant organs and subsequently adapt to the foreign microenvironment. Simulating the physiology of tumor metastatic events in a realistic and three-dimensional (3D) manner is a challenge for in vitro modeling. 3D bioprinting strategies, which can generate well-customized and bionic structures, enable the exploration of dynamic tumor metastasis process in a species-homologous, high-throughput and reproducible way. In this review, we summarize the recent application of 3D bioprinting in constructing in vitro tumor metastatic models and discuss its advantages and current limitations. Further perspectives on how to harness the potential of accessible 3D bioprinting strategies to better model tumor metastasis and guide anti-cancer therapies are also provided.
Collapse
Affiliation(s)
- Manqing Lin
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian 116023, China
| | - Mengyi Tang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian 116023, China
| | - Wenzhe Duan
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian 116023, China
| | - Shengkai Xia
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian 116023, China
| | - Wenwen Liu
- Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian 116023, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital, Dalian Medical University, Dalian 116023, China
- Cancer Translational Medicine Research Center, The Second Hospital, Dalian Medical University, Dalian 116023, China
| |
Collapse
|
7
|
Shao C, Cao T, Wang X, Fan Q, Ye F. Reconstruction of the alveolar-capillary barrier in vitro based on a photo-responsive stretchable Janus membrane. SMART MEDICINE 2023; 2:e20220035. [PMID: 39188563 PMCID: PMC11235665 DOI: 10.1002/smmd.20220035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/05/2023] [Indexed: 08/28/2024]
Abstract
The lung is the respiratory organ of the human body, and the alveoli are the most basic functional units of the lung. Herein, a photo-responsive stretchable Janus membrane was proposed for the reconstruction of the alveolar-capillary barrier in vitro. This Janus membrane was fabricated by photocrosslinking methylacrylamide gelatin (Gelma) hydrogel and N-isoacrylamide (NIPAM) hydrogel mixed with graphene oxide (GO). The Gelma hydrogel containing large amounts of collagen provides a natural extracellular matrix environment for cell growth, while the temperature-sensitive NIPAM hydrogel combined with GO gives the membrane a light-controlled stretching property. Based on this Janus membrane, an open polydimethylsiloxane chip was established to coculture alveolar epithelial cells and vascular endothelial cells at the air-liquid interface. It was demonstrated that the alveolar epithelial cells cultured on the upper side of the Janus membrane could express epithelial cell marker protein E-cadherin and secrete alveolar surfactant. In addition, VE-cadherin, an endothelium-specific protein located at the junction between endothelial cells, was also detected in vascular endothelial cells cultured on the underside of Janus membrane. The constructed lung tissue model with the dynamically stretchable Janus membrane is well-suited for COVID-19 infection studies and drug testing.
Collapse
Affiliation(s)
- Changmin Shao
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Ting Cao
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijingChina
| | - Xiaochen Wang
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijingChina
| | - Qihui Fan
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijingChina
| | - Fangfu Ye
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
- Beijing National Laboratory for Condensed Matter PhysicsInstitute of PhysicsChinese Academy of SciencesBeijingChina
| |
Collapse
|
8
|
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
|
9
|
Ni K, Che B, Yang C, Qin Y, Gu R, Wang C, Luo M, Deng L. Emerging toolset of three-dimensional pulmonary cell culture models for simulating lung pathophysiology towards mechanistic elucidation and therapeutic treatment of SARS-COV-2 infection. Front Pharmacol 2022; 13:1033043. [PMID: 36578545 PMCID: PMC9790924 DOI: 10.3389/fphar.2022.1033043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The ongoing COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) poses a never before seen challenge to human health and the world economy. However, it is difficult to widely use conventional animal and cell culture models in understanding the underlying pathological mechanisms of COVID-19, which in turn hinders the development of relevant therapeutic treatments, including drugs. To overcome this challenge, various three-dimensional (3D) pulmonary cell culture models such as organoids are emerging as an innovative toolset for simulating the pathophysiology occurring in the respiratory system, including bronchial airways, alveoli, capillary network, and pulmonary interstitium, which provide a robust and powerful platform for studying the process and underlying mechanisms of SARS-CoV-2 infection among the potential primary targets in the lung. This review introduces the key features of some of these recently developed tools, including organoid, lung-on-a-chip, and 3D bioprinting, which can recapitulate different structural compartments of the lung and lung function, in particular, accurately resembling the human-relevant pathophysiology of SARS-CoV-2 infection in vivo. In addition, the recent progress in developing organoids for alveolar and airway disease modeling and their applications for discovering drugs against SARS-CoV-2 infection are highlighted. These innovative 3D cell culture models together may hold the promise to fully understand the pathogenesis and eventually eradicate the pandemic of COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou, Jiangsu, China
| |
Collapse
|
10
|
Metelmann IB, Kraemer S, Steinert M, Langer S, Stock P, Kurow O. Novel 3D organotypic co-culture model of pleura. PLoS One 2022; 17:e0276978. [PMID: 36454800 PMCID: PMC9714887 DOI: 10.1371/journal.pone.0276978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Pleural mesothelial cells are the predominant cell type in the pleural cavity, but their role in the pathogenesis of pleural diseases needs to be further elucidated. 3D organotypic models are an encouraging approach for an in vivo understanding of molecular disease development. The aim of the present study was to develop a 3D organotypic model of the pleural mesothelium. Specimens of human pleura parietalis were obtained from patients undergoing surgery at the University Hospital Leipzig, Germany. 3D co-culture model of pleura was established from human pleural mesothelial cells and fibroblasts. The model was compared to human pleura tissue by phase-contrast and light microscopy, immunochemistry and -fluorescence as well as solute permeation test. Histological assessment of the 3D co-culture model displayed the presence of both cell types mimicking the morphology of the human pleura. Vimentin and Cytokeratin, PHD1 showed a similar expression pattern in pleural biopsies and 3D model. Expression of Ki-67 indicates the presence of proliferating cells. Tight junctional marker ZO-1 was found localized at contact zones between mesothelial cells. Each of these markers were expressed in both the 3D co-culture model and human biopsies. Permeability of 3D organotypic co-culture model of pleura was found to be higher for 70 kDa-Dextran and no significant difference was seen in the permeability for small dextran (4 kDa). In summary, the presented 3D organoid of pleura functions as a robust assay for pleural research serving as a precise reproduction of the in vivo morphology and microenvironment.
Collapse
Affiliation(s)
- Isabella B. Metelmann
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Sebastian Kraemer
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Matthias Steinert
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Stefan Langer
- Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Peggy Stock
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Olga Kurow
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
11
|
Humbert MV, Spalluto CM, Bell J, Blume C, Conforti F, Davies ER, Dean LSN, Elkington P, Haitchi HM, Jackson C, Jones MG, Loxham M, Lucas JS, Morgan H, Polak M, Staples KJ, Swindle EJ, Tezera L, Watson A, Wilkinson TMA. Towards an artificial human lung: modelling organ-like complexity to aid mechanistic understanding. Eur Respir J 2022; 60:2200455. [PMID: 35777774 DOI: 10.1183/13993003.00455-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/11/2022] [Indexed: 11/05/2022]
Abstract
Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.
Collapse
Affiliation(s)
- Maria Victoria Humbert
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Cosma Mirella Spalluto
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- M.V. Humbert and C.M. Spalluto are co-first authors and contributed equally to this work
| | - Joseph Bell
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Cornelia Blume
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Franco Conforti
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Elizabeth R Davies
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Lareb S N Dean
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Paul Elkington
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hans Michael Haitchi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Claire Jackson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Mark G Jones
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Matthew Loxham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Hywel Morgan
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Marta Polak
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Karl J Staples
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Emily J Swindle
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Liku Tezera
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Infection and Immunity, Faculty of Medicine, University College London, London, UK
| | - Alastair Watson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Tom M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
12
|
McCloskey MC, Zhang VZ, Ahmad SD, Walker S, Romanick SS, Awad HA, McGrath JL. Sourcing cells for in vitro models of human vascular barriers of inflammation. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:979768. [PMID: 36483299 PMCID: PMC9724237 DOI: 10.3389/fmedt.2022.979768] [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: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 07/20/2023] Open
Abstract
The vascular system plays a critical role in the progression and resolution of inflammation. The contributions of the vascular endothelium to these processes, however, vary with tissue and disease state. Recently, tissue chip models have emerged as promising tools to understand human disease and for the development of personalized medicine approaches. Inclusion of a vascular component within these platforms is critical for properly evaluating most diseases, but many models to date use "generic" endothelial cells, which can preclude the identification of biomedically meaningful pathways and mechanisms. As the knowledge of vascular heterogeneity and immune cell trafficking throughout the body advances, tissue chip models should also advance to incorporate tissue-specific cells where possible. Here, we discuss the known heterogeneity of leukocyte trafficking in vascular beds of some commonly modeled tissues. We comment on the availability of different tissue-specific cell sources for endothelial cells and pericytes, with a focus on stem cell sources for the full realization of personalized medicine. We discuss sources available for the immune cells needed to model inflammatory processes and the findings of tissue chip models that have used the cells to studying transmigration.
Collapse
Affiliation(s)
- Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Victor Z. Zhang
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samuel Walker
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Samantha S. Romanick
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| | - Hani A. Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, United States
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, United States
| |
Collapse
|
13
|
Karra N, Fernandes J, Swindle EJ, Morgan H. Integrating an aerosolized drug delivery device with conventional static cultures and a dynamic airway barrier microphysiological system. BIOMICROFLUIDICS 2022; 16:054102. [PMID: 36118260 PMCID: PMC9473724 DOI: 10.1063/5.0100019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Organ on a chip or microphysiological systems (MPSs) aim to resolve current challenges surrounding drug discovery and development resulting from an unrepresentative static cell culture or animal models that are traditionally used by generating a more physiologically relevant environment. Many different airway MPSs have been developed that mimic alveolar or bronchial interfaces, but few methods for aerosol drug delivery at the air-liquid interface exist. This work demonstrates a compact Surface Acoustic Wave (SAW) drug delivery device that generates an aerosol of respirable size for delivery of compounds directly onto polarized or differentiated epithelial cell cultures within an airway barrier MPS and conventional static inserts. As proof of principle, the SAW drug delivery device was used to nebulize viral dsRNA analog poly I:C and steroids fluticasone and dexamethasone without disrupting their biological function.
Collapse
Affiliation(s)
- Nikita Karra
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, United Kingdom
| | - Joao Fernandes
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, United Kingdom
| | | | - Hywel Morgan
- Author to whom correspondence should be addressed:
| |
Collapse
|
14
|
Nof E, Zidan H, Artzy-Schnirman A, Mouhadeb O, Beckerman M, Bhardwaj S, Elias-Kirma S, Gur D, Beth-Din A, Levenberg S, Korin N, Ordentlich A, Sznitman J. Human Multi-Compartment Airways-on-Chip Platform for Emulating Respiratory Airborne Transmission: From Nose to Pulmonary Acini. Front Physiol 2022; 13:853317. [PMID: 35350687 PMCID: PMC8957966 DOI: 10.3389/fphys.2022.853317] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022] Open
Abstract
The past decade has witnessed tremendous endeavors to deliver novel preclinical in vitro lung models for pulmonary research endpoints, including foremost with the advent of organ- and lung-on-chips. With growing interest in aerosol transmission and infection of respiratory viruses within a host, most notably the SARS-CoV-2 virus amidst the global COVID-19 pandemic, the importance of crosstalk between the different lung regions (i.e., extra-thoracic, conductive and respiratory), with distinct cellular makeups and physiology, are acknowledged to play an important role in the progression of the disease from the initial onset of infection. In the present Methods article, we designed and fabricated to the best of our knowledge the first multi-compartment human airway-on-chip platform to serve as a preclinical in vitro benchmark underlining regional lung crosstalk for viral infection pathways. Combining microfabrication and 3D printing techniques, our platform mimics key elements of the respiratory system spanning (i) nasal passages that serve as the alleged origin of infections, (ii) the mid-bronchial airway region and (iii) the deep acinar region, distinct with alveolated airways. Crosstalk between the three components was exemplified in various assays. First, viral-load (including SARS-CoV-2) injected into the apical partition of the nasal compartment was detected in distal bronchial and acinar components upon applying physiological airflow across the connected compartment models. Secondly, nebulized viral-like dsRNA, poly I:C aerosols were administered to the nasal apical compartment, transmitted to downstream compartments via respiratory airflows and leading to an elevation in inflammatory cytokine levels secreted by distinct epithelial cells in each respective compartment. Overall, our assays establish an in vitro methodology that supports the hypothesis for viral-laden airflow mediated transmission through the respiratory system cellular landscape. With a keen eye for broader end user applications, we share detailed methodologies for fabricating, assembling, calibrating, and using our multi-compartment platform, including open-source fabrication files. Our platform serves as an early proof-of-concept that can be readily designed and adapted to specific preclinical pulmonary research endpoints.
Collapse
Affiliation(s)
- Eliram Nof
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Hikaia Zidan
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Odelia Mouhadeb
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Margarita Beckerman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Saurabh Bhardwaj
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shani Elias-Kirma
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Didi Gur
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Adi Beth-Din
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Netanel Korin
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| | - Arie Ordentlich
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
15
|
de Sá Schiavo Matias G, Carreira ACO, Batista VF, de Carvalho HJC, Miglino MA, Fratini P. In vivo biocompatibility analysis of the recellularized canine tracheal scaffolds with canine epithelial and endothelial progenitor cells. Bioengineered 2022; 13:3551-3565. [PMID: 35109755 PMCID: PMC8974223 DOI: 10.1080/21655979.2021.2020392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Decellularized extracellular matrix (ECM) has frequently been applied as a biomaterial for tissue engineering purposes. When implanted, their role can be essential for partial trachea replacement in patients that require a viable transplant solution. Acellular canine tracheal scaffolds with preserved ECM structure, flexibility, and proteins were obtained by high pressure vacuum decellularization. Here, we aimed to evaluate the cell adhesion and proliferation of canine tracheal epithelial cells (EpC) and canine yolk sac endothelial progenitor cells (YS) cultivated on canine decellularized tracheal scaffolds and test the in vivo biocompatibility of these recellularized scaffolds implanted in BALB-c nude mice. In order to evaluate the recellularization efficiency, scaffolds were evaluated by scanning electron microscopy (SEM), immunofluorescence, DNA quantification, mycoplasma test, and in vivo biocompatibility. The scaffolds sterility was confirmed, and EpC and YS cells were cultured by 7 and 14 days. We demonstrated by SEM, immunofluorescence, and genomic DNA analyzes cell adhesion to tracheal ECM. Then, recellularized scaffolds were in vivo subcutaneously implanted in mice and after 45 days, the fragments were collected and analyzed by Hematoxylin-Eosin and Gömori Trichrome staining and PCNA, CD4, CD8, and CD68 immunohistochemistry. In vivo results confirmed that the implanted tissue remains preserved and proliferative, and no fibrotic tissue process was observed in animals. Finally, our results showed the recellularization success due the preserved ECM proteins, and that these may be suitable to future preclinical studies applications for partial trachea replacement in tissue engineering.
Collapse
Affiliation(s)
- Gustavo de Sá Schiavo Matias
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia O Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Vitória Frias Batista
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Paula Fratini
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Neuromuscular Disease Laboratory, Faculdade de Medicina do ABC (FMABC), Santo André, Brazil
| |
Collapse
|
16
|
Zhang Y, Liu B, Liu Z, Li J. Research progress in synthesis and biological application of quantum dots. NEW J CHEM 2022. [DOI: 10.1039/d2nj02603a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Quantum dots are an excellent choice for biomedical applications due to their special optical properties and quantum confinement effects. This paper reviews the research and application progress of several quantum...
Collapse
|
17
|
Barreiro Carpio M, Dabaghi M, Ungureanu J, Kolb MR, Hirota JA, Moran-Mirabal JM. 3D Bioprinting Strategies, Challenges, and Opportunities to Model the Lung Tissue Microenvironment and Its Function. Front Bioeng Biotechnol 2021; 9:773511. [PMID: 34900964 PMCID: PMC8653950 DOI: 10.3389/fbioe.2021.773511] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Human lungs are organs with an intricate hierarchical structure and complex composition; lungs also present heterogeneous mechanical properties that impose dynamic stress on different tissue components during the process of breathing. These physiological characteristics combined create a system that is challenging to model in vitro. Many efforts have been dedicated to develop reliable models that afford a better understanding of the structure of the lung and to study cell dynamics, disease evolution, and drug pharmacodynamics and pharmacokinetics in the lung. This review presents methodologies used to develop lung tissue models, highlighting their advantages and current limitations, focusing on 3D bioprinting as a promising set of technologies that can address current challenges. 3D bioprinting can be used to create 3D structures that are key to bridging the gap between current cell culture methods and living tissues. Thus, 3D bioprinting can produce lung tissue biomimetics that can be used to develop in vitro models and could eventually produce functional tissue for transplantation. Yet, printing functional synthetic tissues that recreate lung structure and function is still beyond the current capabilities of 3D bioprinting technology. Here, the current state of 3D bioprinting is described with a focus on key strategies that can be used to exploit the potential that this technology has to offer. Despite today's limitations, results show that 3D bioprinting has unexplored potential that may be accessible by optimizing bioink composition and looking at the printing process through a holistic and creative lens.
Collapse
Affiliation(s)
- Mabel Barreiro Carpio
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Mohammadhossein Dabaghi
- Firestone Institute for Respiratory Health, Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Julia Ungureanu
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Martin R. Kolb
- Firestone Institute for Respiratory Health, Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jeremy A. Hirota
- Firestone Institute for Respiratory Health, Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Jose Manuel Moran-Mirabal
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- Centre for Advanced Light Microscopy, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
18
|
Williams AH, Hebert AM, Boehm RC, Huddleston ME, Jenkins MR, Velev OD, Nelson MT. Bioscaffold Stiffness Mediates Aerosolized Nanoparticle Uptake in Lung Epithelial Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50643-50656. [PMID: 34668373 DOI: 10.1021/acsami.1c09701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this study, highly porous, ultrasoft polymeric mats mimicking human tissues were formed from novel polyurethane soft dendritic colloids (PU SDCs). PU SDCs have a unique fibrillar morphology controlled by antisolvent precipitation. When filtered from suspension, PU SDCs form mechanically robust nonwoven mats. The stiffness of the SDC mats can be tuned for physiological relevance. The unique physiochemical characteristics of the PU SDC particles dictate the mechanical properties resulting in tunable elastic moduli ranging from 200 to 800 kPa. The human lung A549 cells cultured on both stiff and soft PU SDC membranes were found to be viable, capable of supporting the air-liquid interface (ALI) cell culture, and maintained barrier integrity. Furthermore, A549 cellular viability and uptake efficiency of aerosolized tannic acid-coated gold nanoparticles (Ta-Au) was found to depend on elastic modulus and culture conditions. Ta-Au nanoparticle uptake was twofold and fourfold greater on soft PU SDCs, when cultured at submerged and ALI conditions, respectively. The significant increase in endocytosed Ta-Au resulted in a 20% decrease in viability, and a 4-fold increase in IL-8 cytokine secretion when cultured on soft PU SDCs at ALI. Common tissue culture materials exhibit super-physiological elastic moduli, a factor found to be critical in analyzing nanomaterial cellular interactions and biological responses.
Collapse
Affiliation(s)
- Austin H Williams
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Adrien M Hebert
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Robert C Boehm
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Mary E Huddleston
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
- UES, Inc., Dayton, Ohio 45432, United States
| | - Meghan R Jenkins
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
- UES, Inc., Dayton, Ohio 45432, United States
| | - Orlin D Velev
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - M Tyler Nelson
- Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
19
|
Young BM, Antczak LAM, Shankar K, Heise RL. A Two-Step Bioreactor for Decellularized Lung Epithelialization. Cells Tissues Organs 2021; 210:301-310. [PMID: 34500450 DOI: 10.1159/000517622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/30/2021] [Indexed: 11/19/2022] Open
Abstract
Bioreactors for the reseeding of decellularized lung scaffolds have evolved with various advancements, including biomimetic mechanical stimulation, constant nutrient flow, multi-output monitoring, and large mammal scaling. Although dynamic bioreactors are not new to the field of lung bioengineering, ideal conditions during cell seeding have not been extensively studied or controlled. To address the lack of cell dispersal in traditional seeding methods, we have designed a two-step bioreactor. The first step is a novel system that rotates a seeded lung every 20 min at different angles in a sequence designed to anchor 20% of cells to a particular location based on the known rate of attachment. The second step involves perfusion-ventilation culture to ensure nutrient dispersion and cellular growth. Compared to statically seeded lungs, rotationally seeded lungs had significantly increased dsDNA content and more uniform cellular distribution after perfusion and ventilation had been administered. The addition of this novel seeding system before traditional culture methods will aid in recellularizing the lung and other geometrically complex organs for tissue engineering.
Collapse
Affiliation(s)
- Bethany M Young
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Leigh-Ann M Antczak
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Keerthana Shankar
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, USA.,Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
20
|
Artzy-Schnirman A, Arber Raviv S, Doppelt Flikshtain O, Shklover J, Korin N, Gross A, Mizrahi B, Schroeder A, Sznitman J. Advanced human-relevant in vitro pulmonary platforms for respiratory therapeutics. Adv Drug Deliv Rev 2021; 176:113901. [PMID: 34331989 PMCID: PMC7611797 DOI: 10.1016/j.addr.2021.113901] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 02/08/2023]
Abstract
Over the past years, advanced in vitro pulmonary platforms have witnessed exciting developments that are pushing beyond traditional preclinical cell culture methods. Here, we discuss ongoing efforts in bridging the gap between in vivo and in vitro interfaces and identify some of the bioengineering challenges that lie ahead in delivering new generations of human-relevant in vitro pulmonary platforms. Notably, in vitro strategies using foremost lung-on-chips and biocompatible "soft" membranes have focused on platforms that emphasize phenotypical endpoints recapitulating key physiological and cellular functions. We review some of the most recent in vitro studies underlining seminal therapeutic screens and translational applications and open our discussion to promising avenues of pulmonary therapeutic exploration focusing on liposomes. Undeniably, there still remains a recognized trade-off between the physiological and biological complexity of these in vitro lung models and their ability to deliver assays with throughput capabilities. The upcoming years are thus anticipated to see further developments in broadening the applicability of such in vitro systems and accelerating therapeutic exploration for drug discovery and translational medicine in treating respiratory disorders.
Collapse
Affiliation(s)
- Arbel Artzy-Schnirman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Sivan Arber Raviv
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | | | - Jeny Shklover
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Adi Gross
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Boaz Mizrahi
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Avi Schroeder
- Department of Chemical, Technion - Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical, Technion - Israel Institute of Technology, 32000 Haifa, Israel.
| |
Collapse
|
21
|
Bennet TJ, Randhawa A, Hua J, Cheung KC. Airway-On-A-Chip: Designs and Applications for Lung Repair and Disease. Cells 2021; 10:1602. [PMID: 34206722 PMCID: PMC8304815 DOI: 10.3390/cells10071602] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/22/2022] Open
Abstract
The lungs are affected by illnesses including asthma, chronic obstructive pulmonary disease, and infections such as influenza and SARS-CoV-2. Physiologically relevant models for respiratory conditions will be essential for new drug development. The composition and structure of the lung extracellular matrix (ECM) plays a major role in the function of the lung tissue and cells. Lung-on-chip models have been developed to address some of the limitations of current two-dimensional in vitro models. In this review, we describe various ECM substitutes utilized for modeling the respiratory system. We explore the application of lung-on-chip models to the study of cigarette smoke and electronic cigarette vapor. We discuss the challenges and opportunities related to model characterization with an emphasis on in situ characterization methods, both established and emerging. We discuss how further advancements in the field, through the incorporation of interstitial cells and ECM, have the potential to provide an effective tool for interrogating lung biology and disease, especially the mechanisms that involve the interstitial elements.
Collapse
Affiliation(s)
- Tanya J. Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Avineet Randhawa
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jessica Hua
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.J.B.); (A.R.); (J.H.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
22
|
Archer F, Bobet-Erny A, Gomes M. State of the art on lung organoids in mammals. Vet Res 2021; 52:77. [PMID: 34078444 PMCID: PMC8170649 DOI: 10.1186/s13567-021-00946-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
The number and severity of diseases affecting lung development and adult respiratory function have stimulated great interest in developing new in vitro models to study lung in different species. Recent breakthroughs in 3-dimensional (3D) organoid cultures have led to new physiological in vitro models that better mimic the lung than conventional 2D cultures. Lung organoids simulate multiple aspects of the real organ, making them promising and useful models for studying organ development, function and disease (infection, cancer, genetic disease). Due to their dynamics in culture, they can serve as a sustainable source of functional cells (biobanking) and be manipulated genetically. Given the differences between species regarding developmental kinetics, the maturation of the lung at birth, the distribution of the different cell populations along the respiratory tract and species barriers for infectious diseases, there is a need for species-specific lung models capable of mimicking mammal lungs as they are of great interest for animal health and production, following the One Health approach. This paper reviews the latest developments in the growing field of lung organoids.
Collapse
Affiliation(s)
- Fabienne Archer
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France.
| | - Alexandra Bobet-Erny
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| | - Maryline Gomes
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| |
Collapse
|
23
|
Nossa R, Costa J, Cacopardo L, Ahluwalia A. Breathing in vitro: Designs and applications of engineered lung models. J Tissue Eng 2021; 12:20417314211008696. [PMID: 33996022 PMCID: PMC8107677 DOI: 10.1177/20417314211008696] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review is to provide a systematic design guideline to users, particularly engineers interested in developing and deploying lung models, and biologists seeking to identify a suitable platform for conducting in vitro experiments involving pulmonary cells or tissues. We first discuss the state of the art on lung in vitro models, describing the most simplistic and traditional ones. Then, we analyze in further detail the more complex dynamic engineered systems that either provide mechanical cues, or allow for more predictive exposure studies, or in some cases even both. This is followed by a dedicated section on microchips of the lung. Lastly, we present a critical discussion of the different characteristics of each type of system and the criteria which may help researchers select the most appropriate technology according to their specific requirements. Readers are encouraged to refer to the tables accompanying the different sections where comprehensive and quantitative information on the operating parameters and performance of the different systems reported in the literature is provided.
Collapse
|
24
|
Ng WL, Ayi TC, Liu YC, Sing SL, Yeong WY, Tan BH. Fabrication and Characterization of 3D Bioprinted Triple-layered Human Alveolar Lung Models. Int J Bioprint 2021; 7:332. [PMID: 33997432 PMCID: PMC8114097 DOI: 10.18063/ijb.v7i2.332] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
The global prevalence of respiratory diseases caused by infectious pathogens has resulted in an increased demand for realistic in-vitro alveolar lung models to serve as suitable disease models. This demand has resulted in the fabrication of numerous two-dimensional (2D) and three-dimensional (3D) in-vitro alveolar lung models. The ability to fabricate these 3D in-vitro alveolar lung models in an automated manner with high repeatability and reliability is important for potential scalable production. In this study, we reported the fabrication of human triple-layered alveolar lung models comprising of human lung epithelial cells, human endothelial cells, and human lung fibroblasts using the drop-on-demand (DOD) 3D bioprinting technique. The polyvinylpyrrolidone-based bio-inks and the use of a 300 mm nozzle diameter improved the repeatability of the bioprinting process by achieving consistent cell output over time using different human alveolar lung cells. The 3D bioprinted human triple-layered alveolar lung models were able to maintain cell viability with relative similar proliferation profile over time as compared to non-printed cells. This DOD 3D bioprinting platform offers an attractive tool for highly repeatable and scalable fabrication of 3D in-vitro human alveolar lung models.
Collapse
Affiliation(s)
- Wei Long Ng
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore.,HP-NTU Digital Manufacturing Corporate Lab, 65 Nanyang Avenue, 637460, Singapore
| | - Teck Choon Ayi
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, 117510, Singapore
| | - Yi-Chun Liu
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, 117510, Singapore
| | - Swee Leong Sing
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore.,HP-NTU Digital Manufacturing Corporate Lab, 65 Nanyang Avenue, 637460, Singapore
| | - Boon-Huan Tan
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 27 Medical Drive, 117510, Singapore.,Lee Kong Chian School of Medicine, Novena Campus, Nanyang Technological University, 11 Mandalay Road, 308232, Singapore
| |
Collapse
|
25
|
Barosova H, Karakocak BB, Septiadi D, Petri-Fink A, Stone V, Rothen-Rutishauser B. An In Vitro Lung System to Assess the Proinflammatory Hazard of Carbon Nanotube Aerosols. Int J Mol Sci 2020; 21:ijms21155335. [PMID: 32727099 PMCID: PMC7432093 DOI: 10.3390/ijms21155335] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
In vitro three-dimensional (3D) lung cell models have been thoroughly investigated in recent years and provide a reliable tool to assess the hazard associated with nanomaterials (NMs) released into the air. In this study, a 3D lung co-culture model was optimized to assess the hazard potential of multiwalled carbon nanotubes (MWCNTs), which is known to provoke inflammation and fibrosis, critical adverse outcomes linked to acute and prolonged NM exposure. The lung co-cultures were exposed to MWCNTs at the air-liquid interface (ALI) using the VITROCELL® Cloud system while considering realistic occupational exposure doses. The co-culture model was composed of three human cell lines: alveolar epithelial cells (A549), fibroblasts (MRC-5), and macrophages (differentiated THP-1). The model was exposed to two types of MWCNTs (Mitsui-7 and Nanocyl) at different concentrations (2–10 μg/cm2) to assess the proinflammatory as well as the profibrotic responses after acute (24 h, one exposure) and prolonged (96 h, repeated exposures) exposure cycles. The results showed that acute or prolonged exposure to different concentrations of the tested MWCNTs did not induce cytotoxicity or apparent profibrotic response; however, suggested the onset of proinflammatory response.
Collapse
Affiliation(s)
- Hana Barosova
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland; (H.B.); (B.B.K.); (D.S.); (A.P.-F.)
- Institute of Experimental Medicine of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Bedia Begum Karakocak
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland; (H.B.); (B.B.K.); (D.S.); (A.P.-F.)
| | - Dedy Septiadi
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland; (H.B.); (B.B.K.); (D.S.); (A.P.-F.)
| | - Alke Petri-Fink
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland; (H.B.); (B.B.K.); (D.S.); (A.P.-F.)
- Department of Chemistry, University of Fribourg, 1700 Fribourg, Switzerland
| | - Vicki Stone
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, UK;
| | - Barbara Rothen-Rutishauser
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, 1700 Fribourg, Switzerland; (H.B.); (B.B.K.); (D.S.); (A.P.-F.)
- Correspondence: ; Tel.: +41-26-300-9502
| |
Collapse
|
26
|
Movia D, Prina-Mello A. Preclinical Development of Orally Inhaled Drugs (OIDs)-Are Animal Models Predictive or Shall We Move Towards In Vitro Non-Animal Models? Animals (Basel) 2020; 10:E1259. [PMID: 32722259 PMCID: PMC7460012 DOI: 10.3390/ani10081259] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
Respiratory diseases constitute a huge burden in our society, and the global respiratory drug market currently grows at an annual rate between 4% and 6%. Inhalation is the preferred administration method for treating respiratory diseases, as it: (i) delivers the drug directly at the site of action, resulting in a rapid onset; (ii) is painless, thus improving patients' compliance; and (iii) avoids first-pass metabolism reducing systemic side effects. Inhalation occurs through the mouth, with the drug generally exerting its therapeutic action in the lungs. In the most recent years, orally inhaled drugs (OIDs) have found application also in the treatment of systemic diseases. OIDs development, however, currently suffers of an overall attrition rate of around 70%, meaning that seven out of 10 new drug candidates fail to reach the clinic. Our commentary focuses on the reasons behind the poor OIDs translation into clinical products for the treatment of respiratory and systemic diseases, with particular emphasis on the parameters affecting the predictive value of animal preclinical tests. We then review the current advances in overcoming the limitation of animal animal-based studies through the development and adoption of in vitro, cell-based new approach methodologies (NAMs).
Collapse
Affiliation(s)
- Dania Movia
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College, The University of Dublin, Dublin D8, Ireland;
| | - Adriele Prina-Mello
- Laboratory for Biological Characterisation of Advanced Materials (LBCAM), Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College, The University of Dublin, Dublin D8, Ireland;
- AMBER Centre, CRANN Institute, Trinity College, The University of Dublin, Dublin D2, Ireland
| |
Collapse
|
27
|
Figliuzzi M, Tironi M, Longaretti L, Mancini A, Teoldi F, Sangalli F, Remuzzi A. Copper-dependent biological effects of particulate matter produced by brake systems on lung alveolar cells. Arch Toxicol 2020; 94:2965-2979. [PMID: 32577786 DOI: 10.1007/s00204-020-02812-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Road traffic is one of the main sources of particulate emissions into the environment and has an increasing, negative impact on the release of potentially dangerous materials. Vehicle brakes release a significant amount of wear particles, and knowledge regarding their possible adverse effects is limited. One of the most dangerous elements contained in brake pads is copper (Cu), known to be toxic for human health. Therefore, our aim was to study the cell toxicity of particulate matter (PM) produced by different combinations of braking discs and pads containing different amounts of Cu. We investigated whether brake-derived microparticles have toxic effects on lung cells proportionally to their Cu content. Analyte content was measured in friction materials by XRFS and in PM2.5 captured during braking tests using SEM/EDX. The biological impact of brake-derived PM2.5 was investigated on a human epithelial alveolar cell line (A549). Cell viability, oxidative stress, mitochondrial membrane potential, apoptosis, and the pro-inflammatory response of the cells, as well as gene expression, were assessed following exposure to increasing PM2.5 concentrations (1, 10, 100, 200, and 500 µg/ml). The brake debris with the lowest Cu content did not induce significant changes in biological effects on A549 cells compared to normal controls, except for ROS production and IL6 gene expression. PM2.5 containing higher Cu quantities induced cell toxicity that correlated with Cu concentration. Our data suggest that the toxicity of PM2.5 from the brake system is mainly related to Cu content, thus confirming that eliminating Cu from brake pads will be beneficial for human health in urbanized environments.
Collapse
Affiliation(s)
- Marina Figliuzzi
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy.
| | - Matteo Tironi
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy
| | - Lorena Longaretti
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, Bergamo, Italy
| | - Alessandro Mancini
- Laboratorio Materiali Advanced R&D Brembo S.P.A, Viale Europa, 2, Stezzano, BG, Italy
| | - Federico Teoldi
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Negri 2, Milan, Italy
| | - Fabio Sangalli
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Stezzano 87, 24126, Bergamo, Italy
| | - Andrea Remuzzi
- Department of Management, Information and Production Engineering, University of Bergamo, Viale Marconi 5, Dalmine, BG, Italy
| |
Collapse
|
28
|
Yanagihara T, Chong SG, Vierhout M, Hirota JA, Ask K, Kolb M. Current models of pulmonary fibrosis for future drug discovery efforts. Expert Opin Drug Discov 2020; 15:931-941. [PMID: 32396021 DOI: 10.1080/17460441.2020.1755252] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Pulmonary fibrosis includes several lung disorders characterized by progressive fibrosis, of which idiopathic pulmonary fibrosis (IPF) is a particularly severe form with a median survival time of 3-5 years after diagnosis. Although numerous compounds have shown efficacy in attenuating pulmonary fibrosis using animal models, only a few compounds have shown their beneficial effects for IPF in clinical trials. Thus, there is an emergent need to improve the preclinical development process to better identify, characterize and select clinically useful targets. AREAS COVERED In this review, the authors extensively describe current models of pulmonary fibrosis, including rodent models, ex vivo models, and in vitro models. EXPERT OPINION Based upon our current understanding, improving the identification and characterization of clinically relevant molecules or pathways responsible for progressive fibrotic diseases and use of the appropriate preclinical model system to test these will likely be required to improve the drug development pipeline for pulmonary fibrosis. Combination with appropriate preclinical models with ex vivo (precision-cut lung slices) or in vitro models would be beneficial for high-throughput drug discovery or validation of drug effects.
Collapse
Affiliation(s)
- Toyoshi Yanagihara
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada.,Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University , Fukuoka, Japan
| | - Sy Giin Chong
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Megan Vierhout
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Jeremy A Hirota
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| |
Collapse
|
29
|
Barosova H, Maione AG, Septiadi D, Sharma M, Haeni L, Balog S, O'Connell O, Jackson GR, Brown D, Clippinger AJ, Hayden P, Petri-Fink A, Stone V, Rothen-Rutishauser B. Use of EpiAlveolar Lung Model to Predict Fibrotic Potential of Multiwalled Carbon Nanotubes. ACS NANO 2020; 14:3941-3956. [PMID: 32167743 DOI: 10.1021/acsnano.9b06860] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Expansion in production and commercial use of nanomaterials increases the potential human exposure during the lifecycle of these materials (production, use, and disposal). Inhalation is a primary route of exposure to nanomaterials; therefore it is critical to assess their potential respiratory hazard. Herein, we developed a three-dimensional alveolar model (EpiAlveolar) consisting of human primary alveolar epithelial cells, fibroblasts, and endothelial cells, with or without macrophages for predicting long-term responses to aerosols. Following thorough characterization of the model, proinflammatory and profibrotic responses based on the adverse outcome pathway concept for lung fibrosis were assessed upon repeated subchronic exposures (up to 21 days) to two types of multiwalled carbon nanotubes (MWCNTs) and silica quartz particles. We simulate occupational exposure doses for the MWCNTs (1-30 μg/cm2) using an air-liquid interface exposure device (VITROCELL Cloud) with repeated exposures over 3 weeks. Specific key events leading to lung fibrosis, such as barrier integrity and release of proinflammatory and profibrotic markers, show the responsiveness of the model. Nanocyl induced, in general, a less pronounced reaction than Mitsui-7, and the cultures with human monocyte-derived macrophages (MDMs) showed the proinflammatory response at later time points than those without MDMs. In conclusion, we present a robust alveolar model to predict inflammatory and fibrotic responses upon exposure to MWCNTs.
Collapse
Affiliation(s)
- Hana Barosova
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Anna G Maione
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts 01721, United States
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Monita Sharma
- PETA International Science Consortium Ltd., 8 All Saints Street, London N1 9RL, U.K
| | - Laetitia Haeni
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Olivia O'Connell
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts 01721, United States
| | - George R Jackson
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts 01721, United States
| | - David Brown
- Nano-Safety Research Group, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | - Amy J Clippinger
- PETA International Science Consortium Ltd., 8 All Saints Street, London N1 9RL, U.K
| | - Patrick Hayden
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts 01721, United States
- BioSurfaces, Inc., 200 Homer Ave, Ashland, Massachusetts 01721, United States
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Vicki Stone
- Nano-Safety Research Group, Heriot-Watt University, Edinburgh EH14 4AS, U.K
| | | |
Collapse
|
30
|
Dorrello NV, Vunjak-Novakovic G. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche. Front Bioeng Biotechnol 2020; 8:269. [PMID: 32351946 PMCID: PMC7174601 DOI: 10.3389/fbioe.2020.00269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
Collapse
Affiliation(s)
- N Valerio Dorrello
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
31
|
Ehrmann S, Schmid O, Darquenne C, Rothen-Rutishauser B, Sznitman J, Yang L, Barosova H, Vecellio L, Mitchell J, Heuze-Vourc’h N. Innovative preclinical models for pulmonary drug delivery research. Expert Opin Drug Deliv 2020; 17:463-478. [PMID: 32057260 PMCID: PMC8083945 DOI: 10.1080/17425247.2020.1730807] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/11/2020] [Indexed: 02/08/2023]
Abstract
Introduction: Pulmonary drug delivery is a complex field of research combining physics which drive aerosol transport and deposition and biology which underpins efficacy and toxicity of inhaled drugs. A myriad of preclinical methods, ranging from in-silico to in-vitro, ex-vivo and in-vivo, can be implemented.Areas covered: The present review covers in-silico mathematical and computational fluid dynamics modelization of aerosol deposition, cascade impactor technology to estimated drug delivery and deposition, advanced in-vitro cell culture methods and associated aerosol exposure, lung-on-chip technology, ex-vivo modeling, in-vivo inhaled drug delivery, lung imaging, and longitudinal pharmacokinetic analysis.Expert opinion: No single preclinical model can be advocated; all methods are fundamentally complementary and should be implemented based on benefits and drawbacks to answer specific scientific questions. The overall best scientific strategy depends, among others, on the product under investigations, inhalation device design, disease of interest, clinical patient population, previous knowledge. Preclinical testing is not to be separated from clinical evaluation, as small proof-of-concept clinical studies or conversely large-scale clinical big data may inform preclinical testing. The extend of expertise required for such translational research is unlikely to be found in one single laboratory calling for the setup of multinational large-scale research consortiums.
Collapse
Affiliation(s)
- Stephan Ehrmann
- CHRU Tours, Médecine Intensive Réanimation, CIC INSERM 1415, CRICS-TriggerSep network, Tours France
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München – German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Chantal Darquenne
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, MC0623A, La Jolla, CA 92093-0623, United States
| | | | - Josue Sznitman
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Julius Silver building, Office 246, Haifa 32000, Israel
| | - Lin Yang
- Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), Max-Lebsche-Platz 31, 81377 Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München – German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Hana Barosova
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg, Switzerland
| | - Laurent Vecellio
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| | - Jolyon Mitchell
- Jolyon Mitchell Inhaler Consulting Services Inc., 1154 St. Anthony Road, London, Ontario, Canada, N6H 2R1
| | - Nathalie Heuze-Vourc’h
- INSERM, Centre d’étude des pathologies respiratoires, U1100, Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
32
|
Ainslie GR, Davis M, Ewart L, Lieberman LA, Rowlands DJ, Thorley AJ, Yoder G, Ryan AM. Microphysiological lung models to evaluate the safety of new pharmaceutical modalities: a biopharmaceutical perspective. LAB ON A CHIP 2019; 19:3152-3161. [PMID: 31469131 DOI: 10.1039/c9lc00492k] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The lung is a complex organ; it is both the initial barrier for inhaled agents and the site of metabolism and therapeutic effect for a subset of systemically administered drugs. Comprised of more than 40 cell types that are responsible for various important functions, the lung's complexity contributes to the subsequent challenges in developing complex in vitro co-culture models (also called microphysiological systems (MPS), complex in vitro models or organs-on-a-chip). Although there are multiple considerations and limitations in the development and qualification of such in vitro systems, MPS exhibit great promise in the fields of pharmacology and toxicology. Successful development and implementation of MPS models may enable mechanistic bridging between non-clinical species and humans, and increase clinical relevance of safety endpoints, while decreasing overall animal use. This article summarizes, from a biopharmaceutical industry perspective, essential elements for the development and qualification of lung MPS models. Its purpose is to guide MPS developers and manufacturers to expedite MPS utilization for safety assessment in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Garrett R Ainslie
- Drug Metabolism and Pharmacokinetics, Theravance Biopharma US, Inc, South San Francisco, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Tebyanian H, Karami A, Nourani MR, Motavallian E, Barkhordari A, Yazdanian M, Seifalian A. Lung tissue engineering: An update. J Cell Physiol 2019; 234:19256-19270. [PMID: 30972749 DOI: 10.1002/jcp.28558] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022]
Abstract
Pulmonary disease is a worldwide public health problem that reduces the life quality and increases the need for hospital admissions as well as the risk of premature death. A common problem is the significant shortage of lungs for transplantation as well as patients must also take immunosuppressive drugs for the rest of their lives to keep the immune system from attacking transplanted organs. Recently, a new strategy has been proposed in the cellular engineering of lung tissue as decellularization approaches. The main components for the lung tissue engineering are: (1) A suitable biological or synthetic three-dimensional (3D) scaffold, (2) source of stem cells or cells, (3) growth factors required to drive cell differentiation and proliferation, and (4) bioreactor, a system that supports a 3D composite biologically active. Although a number of synthetic as well biological 3D scaffold suggested for lung tissue engineering, the current favorite scaffold is decellularized extracellular matrix scaffold. There are a large number of commercial and academic made bioreactors, the favor has been, the one easy to sterilize, physiologically stimuli and support active cell growth as well as clinically translational. The challenges would be to develop a functional lung will depend on the endothelialized microvascular network and alveolar-capillary surface area to exchange gas. A critical review of the each components of lung tissue engineering is presented, following an appraisal of the literature in the last 5 years. This is a multibillion dollar industry and consider unmet clinical need.
Collapse
Affiliation(s)
- Hamid Tebyanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Karami
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Nourani
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ebrahim Motavallian
- Department of General Surgery, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Aref Barkhordari
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre (Ltd), The London Bioscience Innovation Centre, London, UK
| |
Collapse
|
34
|
Czekala L, Simms L, Stevenson M, Tschierske N, Maione AG, Walele T. Toxicological comparison of cigarette smoke and e-cigarette aerosol using a 3D in vitro human respiratory model. Regul Toxicol Pharmacol 2019; 103:314-324. [DOI: 10.1016/j.yrtph.2019.01.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
|
35
|
Hilton G, Barosova H, Petri-Fink A, Rothen-Rutishauser B, Bereman M. Leveraging proteomics to compare submerged versus air-liquid interface carbon nanotube exposure to a 3D lung cell model. Toxicol In Vitro 2019; 54:58-66. [DOI: 10.1016/j.tiv.2018.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/29/2018] [Accepted: 09/17/2018] [Indexed: 01/23/2023]
|
36
|
Kendall LV, Owiny JR, Dohm ED, Knapek KJ, Lee ES, Kopanke JH, Fink M, Hansen SA, Ayers JD. Replacement, Refinement, and Reduction in Animal Studies With Biohazardous Agents. ILAR J 2019; 59:177-194. [DOI: 10.1093/ilar/ily021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Abstract
Animal models are critical to the advancement of our knowledge of infectious disease pathogenesis, diagnostics, therapeutics, and prevention strategies. The use of animal models requires thoughtful consideration for their well-being, as infections can significantly impact the general health of an animal and impair their welfare. Application of the 3Rs—replacement, refinement, and reduction—to animal models using biohazardous agents can improve the scientific merit and animal welfare. Replacement of animal models can use in vitro techniques such as cell culture systems, mathematical models, and engineered tissues or invertebrate animal hosts such as amoeba, worms, fruit flies, and cockroaches. Refinements can use a variety of techniques to more closely monitor the course of disease. These include the use of biomarkers, body temperature, behavioral observations, and clinical scoring systems. Reduction is possible using advanced technologies such as in vivo telemetry and imaging, allowing longitudinal assessment of animals during the course of disease. While there is no single method to universally replace, refine, or reduce animal models, the alternatives and techniques discussed are broadly applicable and they should be considered when infectious disease animal models are developed.
Collapse
Affiliation(s)
- Lon V Kendall
- Department of Microbiology, Immunology and Pathology, and Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| | - James R Owiny
- Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| | - Erik D Dohm
- Animal Resources Program, University of Alabama, Birmingham, Alabama
| | - Katie J Knapek
- Comparative Medicine Training Program, Colorado State University, Fort Collins, Colorado
| | - Erin S Lee
- Animal Resource Center, University of Texas Medical Branch, Galveston, Texas
| | - Jennifer H Kopanke
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado
| | - Michael Fink
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Sarah A Hansen
- Office of Animal Resources, University of Iowa, Iowa City, Iowa
| | - Jessica D Ayers
- Laboratory Animal Resources, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
37
|
Weinhart M, Hocke A, Hippenstiel S, Kurreck J, Hedtrich S. 3D organ models-Revolution in pharmacological research? Pharmacol Res 2019; 139:446-451. [PMID: 30395949 PMCID: PMC7129286 DOI: 10.1016/j.phrs.2018.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/30/2018] [Accepted: 11/01/2018] [Indexed: 01/15/2023]
Abstract
3D organ models have gained increasing attention as novel preclinical test systems and alternatives to animal testing. Over the years, many excellent in vitro tissue models have been developed. In parallel, microfluidic organ-on-a-chip tissue cultures have gained increasing interest for their ability to house several organ models on a single device and interlink these within a human-like environment. In contrast to these advancements, the development of human disease models is still in its infancy. Although major advances have recently been made, efforts still need to be intensified. Human disease models have proven valuable for their ability to closely mimic disease patterns in vitro, permitting the study of pathophysiological features and new treatment options. Although animal studies remain the gold standard for preclinical testing, they have major drawbacks such as high cost and ongoing controversy over their predictive value for several human conditions. Moreover, there is growing political and social pressure to develop alternatives to animal models, clearly promoting the search for valid, cost-efficient and easy-to-handle systems lacking interspecies-related differences. In this review, we discuss the current state of the art regarding 3D organ as well as the opportunities, limitations and future implications of their use.
Collapse
Affiliation(s)
- Marie Weinhart
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Andreas Hocke
- Dept. of Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Stefan Hippenstiel
- Dept. of Infectious and Respiratory Diseases, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Jens Kurreck
- Technical University Berlin, Institute for Biotechnology, Berlin, Germany
| | - Sarah Hedtrich
- Freie Universität Berlin, Institute for Pharmacy, Pharmacology & Toxicology, Königin-Luise-Str. 2-4, Berlin, 14195, Germany.
| |
Collapse
|
38
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|
39
|
Tenenbaum-Katan J, Artzy-Schnirman A, Fishler R, Korin N, Sznitman J. Biomimetics of the pulmonary environment in vitro: A microfluidics perspective. BIOMICROFLUIDICS 2018; 12:042209. [PMID: 29887933 PMCID: PMC5973897 DOI: 10.1063/1.5023034] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 03/20/2018] [Indexed: 05/08/2023]
Abstract
The entire luminal surface of the lungs is populated with a complex yet confluent, uninterrupted airway epithelium in conjunction with an extracellular liquid lining layer that creates the air-liquid interface (ALI), a critical feature of healthy lungs. Motivated by lung disease modelling, cytotoxicity studies, and drug delivery assessments amongst other, in vitro setups have been traditionally conducted using macroscopic cultures of isolated airway cells under submerged conditions or instead using transwell inserts with permeable membranes to model the ALI architecture. Yet, such strategies continue to fall short of delivering a sufficiently realistic physiological in vitro airway environment that cohesively integrates at true-scale three essential pillars: morphological constraints (i.e., airway anatomy), physiological conditions (e.g., respiratory airflows), and biological functionality (e.g., cellular makeup). With the advent of microfluidic lung-on-chips, there have been tremendous efforts towards designing biomimetic airway models of the epithelial barrier, including the ALI, and leveraging such in vitro scaffolds as a gateway for pulmonary disease modelling and drug screening assays. Here, we review in vitro platforms mimicking the pulmonary environment and identify ongoing challenges in reconstituting accurate biological airway barriers that still widely prevent microfluidic systems from delivering mainstream assays for the end-user, as compared to macroscale in vitro cell cultures. We further discuss existing hurdles in scaling up current lung-on-chip designs, from single airway models to more physiologically realistic airway environments that are anticipated to deliver increasingly meaningful whole-organ functions, with an outlook on translational and precision medicine.
Collapse
Affiliation(s)
- Janna Tenenbaum-Katan
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Arbel Artzy-Schnirman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Rami Fishler
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Netanel Korin
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion–Israel Institute of Technology, 32000 Haifa, Israel
| |
Collapse
|
40
|
Barosova H, Chortarea S, Peikertova P, Clift MJD, Petri-Fink A, Kukutschova J, Rothen-Rutishauser B. Biological response of an in vitro human 3D lung cell model exposed to brake wear debris varies based on brake pad formulation. Arch Toxicol 2018; 92:2339-2351. [PMID: 29748788 PMCID: PMC6015608 DOI: 10.1007/s00204-018-2218-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 05/03/2018] [Indexed: 10/25/2022]
Abstract
Wear particles from automotive friction brake pads of various sizes, morphology, and chemical composition are significant contributors towards particulate matter. Knowledge concerning the potential adverse effects following inhalation exposure to brake wear debris is limited. Our aim was, therefore, to generate brake wear particles released from commercial low-metallic and non-asbestos organic automotive brake pads used in mid-size passenger cars by a full-scale brake dynamometer with an environmental chamber simulating urban driving and to deduce their potential hazard in vitro. The collected fractions were analysed using scanning electron microscopy via energy-dispersive X-ray spectroscopy (SEM-EDS) and Raman microspectroscopy. The biological impact of the samples was investigated using a human 3D multicellular model consisting of human epithelial cells (A549) and human primary immune cells (macrophages and dendritic cells) mimicking the human epithelial tissue barrier. The viability, morphology, oxidative stress, and (pro-)inflammatory response of the cells were assessed following 24 h exposure to ~ 12, ~ 24, and ~ 48 µg/cm2 of non-airborne samples and to ~ 3.7 µg/cm2 of different brake wear size fractions (2-4, 1-2, and 0.25-1 µm) applying a pseudo-air-liquid interface approach. Brake wear debris with low-metallic formula does not induce any adverse biological effects to the in vitro lung multicellular model. Brake wear particles from non-asbestos organic formulated pads, however, induced increased (pro-)inflammatory mediator release from the same in vitro system. The latter finding can be attributed to the different particle compositions, specifically the presence of anatase.
Collapse
Affiliation(s)
- Hana Barosova
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Nanotechnology Centre, VŠB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Savvina Chortarea
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Laboratory for Materials-Biology Interactions, Empa, Swiss Federal Laboratories for Materials, Science and Technology, St. Gallen, Switzerland
| | - Pavlina Peikertova
- Nanotechnology Centre, VŠB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Martin J. D. Clift
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- In Vitro Toxicology Group, Swansea University Medical School, Swansea, Wales UK
| | - Alke Petri-Fink
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
- Chemistry Department, University of Fribourg, Fribourg, Switzerland
| | - Jana Kukutschova
- Nanotechnology Centre, VŠB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Barbara Rothen-Rutishauser
- BioNanomaterials Group, Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| |
Collapse
|
41
|
Xue D, Wang Y, Zhang J, Mei D, Wang Y, Chen S. Projection-Based 3D Printing of Cell Patterning Scaffolds with Multiscale Channels. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19428-19435. [PMID: 29782142 DOI: 10.1021/acsami.8b03867] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To fully actualize artificial, cell-laden biological models in tissue engineering, such as 3D organoids and organs-on-a-chip systems, cells need to be patterned such that they can precisely mimic natural microenvironments in vitro. Despite increasing interest in this area, patterning cells at multiscale (∼10 μm to 10 mm) remains a significant challenge in bioengineering. Here, we report a projection-based 3D printing system that achieves rapid and high-resolution fabrication of hydrogel scaffolds featuring intricate channels for multiscale cell patterning. Using this system, we were able to use biocompatible poly(ethylene glycol)diacrylate in fabricating a variety of scaffold architectures, ranging from regular geometries such as serpentine, spiral, and fractal-like to more irregular/intricate geometries, such as biomimetic arborescent and capillary networks. A red food dye solution was able to freely fill all channels in the scaffolds, from the trunk (>1100 μm in width) to the small branch (∼17 μm in width) without an external pump. The dimensions of the printed scaffolds remained stable over 3 days while being immersed in Dulbecco's phosphate-buffered saline at 37 °C, and a penetration analysis revealed that these scaffolds are suitable for metabolic and nutrient transport. Cell patterning experiments showed that red fluorescent protein-transfected A549 human nonsmall lung cancer cells adhered well in the scaffolds' channels, and showed further attachment and penetration during cell culture proliferation.
Collapse
Affiliation(s)
- Dai Xue
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| | | | - Jiaxin Zhang
- Department of Toxicology , Fourth Military Medical University , Xi'an 710032 , China
| | | | | | - Shaochen Chen
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| |
Collapse
|
42
|
Eenjes E, Mertens TCJ, Buscop-van Kempen MJ, van Wijck Y, Taube C, Rottier RJ, Hiemstra PS. A novel method for expansion and differentiation of mouse tracheal epithelial cells in culture. Sci Rep 2018; 8:7349. [PMID: 29743551 PMCID: PMC5943313 DOI: 10.1038/s41598-018-25799-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 04/25/2018] [Indexed: 12/12/2022] Open
Abstract
Air-liquid interface (ALI) cultures of mouse tracheal epithelial cells (MTEC) are a well-established model to study airway epithelial cells, but current methods require large numbers of animals which is unwanted in view of the 3R principle and introduces variation. Moreover, stringent breeding schemes are frequently needed to generate sufficient numbers of genetically modified animals. Current protocols do not incorporate expansion of MTEC, and therefore we developed a protocol to expand MTEC while maintaining their differentiation capacity. MTEC were isolated and expanded using the ROCK inhibitor Y-27632 in presence or absence of the γ-secretase inhibitor DAPT, a Notch pathway inhibitor. Whereas MTEC proliferated without DAPT, growth rate and cell morphology improved in presence of DAPT. ALI-induced differentiation of expanded MTEC resulted in an altered capacity of basal cells to differentiate into ciliated cells, whereas IL-13-induced goblet cell differentiation remained unaffected. Ciliated cell differentiation improved by prolonging the ALI differentiation or by adding DAPT, suggesting that basal cells retain their ability to differentiate. This technique using expansion of MTEC and subsequent ALI differentiation drastically reduces animal numbers and costs for in vitro experiments, and will reduce biological variation. Additionally, we provide novel insights in the dynamics of basal cell populations in vitro.
Collapse
Affiliation(s)
- Evelien Eenjes
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Tinne C J Mertens
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Marjon J Buscop-van Kempen
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Yolanda van Wijck
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Pulmonary Medicine, West German Lung Center, Essen University Hospital, Ruhrlandklinik, University Duisburg-Essen, Essen, Germany
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
43
|
Liu XJ, Hu SW, Xu BY, Zhao G, Li X, Xie FW, Xu JJ, Chen HY. Microfluidic liquid-air dual-gradient chip for synergic effect bio-evaluation of air pollutant. Talanta 2018; 182:202-209. [DOI: 10.1016/j.talanta.2018.01.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/25/2018] [Accepted: 01/30/2018] [Indexed: 10/18/2022]
|
44
|
Effect of off-plane bifurcation angles of primary bronchi on expiratory flows in the human trachea. Comput Biol Med 2018; 95:63-74. [DOI: 10.1016/j.compbiomed.2018.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/17/2018] [Accepted: 01/30/2018] [Indexed: 11/20/2022]
|
45
|
Sundarakrishnan A, Chen Y, Black LD, Aldridge BB, Kaplan DL. Engineered cell and tissue models of pulmonary fibrosis. Adv Drug Deliv Rev 2018; 129:78-94. [PMID: 29269274 DOI: 10.1016/j.addr.2017.12.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/15/2017] [Accepted: 12/16/2017] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis includes several lung disorders characterized by scar formation and Idiopathic Pulmonary Fibrosis (IPF) is a particularly severe form of pulmonary fibrosis of unknown etiology with a mean life expectancy of 3years' post-diagnosis. Treatments for IPF are limited to two FDA approved drugs, pirfenidone and nintedanib. Most lead candidate drugs that are identified in pre-clinical animal studies fail in human clinical trials. Thus, there is a need for advanced humanized in vitro models of the lung to improve candidate treatments prior to moving to human clinical trials. The development of 3D tissue models has created systems capable of emulating human lung structure, function, and cell and matrix interactions. The specific models accomplish these features and preliminary studies conducted using some of these systems have shown potential for in vitro anti-fibrotic drug testing. Further characterization and improvements will enable these tissue models to extend their utility for in vitro drug testing, to help identify signaling pathways and mechanisms for new drug targets, and potentially reduce animal models as standard pre-clinical models of study. In the current review, we contrast different in vitro models based on increasing dimensionality (2D, 2.5D and 3D), with added focus on contemporary 3D pulmonary models of fibrosis.
Collapse
Affiliation(s)
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States; Department of Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
| | - Bree B Aldridge
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States; Department of Molecular Biology & Microbiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States.
| |
Collapse
|
46
|
Löwa A, Jevtić M, Gorreja F, Hedtrich S. Alternatives to animal testing in basic and preclinical research of atopic dermatitis. Exp Dermatol 2018; 27:476-483. [PMID: 29356091 DOI: 10.1111/exd.13498] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2018] [Indexed: 12/29/2022]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease of increasing prevalence, especially in industrialized countries. Roughly 25% of the children and 1%-3% of adults are affected. Although significant progress has been made in the understanding of the pathogenesis of AD, many aspects remain poorly understood. Moreover, there is a pressing need for improved therapeutic options. Studies to elucidate the pathophysiological pathways of AD and to identify novel therapeutic targets over the last few decades have been conducted almost exclusively in animal models. However, in vitro approaches such as 3D skin disease models have recently emerged due to an increasing awareness of distinct interspecies-related differences that hamper the effective translation of results from animal models to humans. In addition, there is growing political and social pressure to develop alternatives to animal models according to the 3Rs principle (reduction, refinement and replacement of animal models).
Collapse
Affiliation(s)
- Anna Löwa
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Marijana Jevtić
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Frida Gorreja
- Örebro University, School of Health and Medical Sciences, Örebro University, Orebro, Sweden
| | - Sarah Hedtrich
- Institute for Pharmacy, Pharmacology & Toxicology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
47
|
Young BM, Shankar K, Allen BP, Pouliot RA, Schneck MB, Mikhaiel NS, Heise RL. Electrospun Decellularized Lung Matrix Scaffold for Airway Smooth Muscle Culture. ACS Biomater Sci Eng 2017; 3:3480-3492. [DOI: 10.1021/acsbiomaterials.7b00384] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Bethany M. Young
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Keerthana Shankar
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Brittany P. Allen
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Robert A. Pouliot
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Matthew B. Schneck
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Nabil S. Mikhaiel
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
| | - Rebecca L. Heise
- Department
of Biomedical Engineering, Virginia Commonwealth University, 800 E. Leigh
Street, Room 1071, Richmond, Virginia 23219, United States
- Department
of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, Virginia 23298, United States
| |
Collapse
|
48
|
Cyr KJ, Avaldi OM, Wikswo JP. Circadian hormone control in a human-on-a-chip: In vitro biology's ignored component? Exp Biol Med (Maywood) 2017; 242:1714-1731. [PMID: 29065796 PMCID: PMC5832251 DOI: 10.1177/1535370217732766] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Organs-on-Chips (OoCs) are poised to reshape dramatically the study of biology by replicating in vivo the function of individual and coupled human organs. Such microphysiological systems (MPS) have already recreated complex physiological responses necessary to simulate human organ function not evident in two-dimensional in vitro biological experiments. OoC researchers hope to streamline pharmaceutical development, accelerate toxicology studies, limit animal testing, and provide new insights beyond the capability of current biological models. However, to develop a physiologically accurate Human-on-a-Chip, i.e., an MPS homunculus that functions as an interconnected, whole-body, model organ system, one must couple individual OoCs with proper fluidic and metabolic scaling. This will enable the study of the effects of organ-organ interactions on the metabolism of drugs and toxins. Critical to these efforts will be the recapitulation of the complex physiological signals that regulate the endocrine, metabolic, and digestive systems. To date, with the exception of research focused on reproductive organs on chips, most OoC research ignores homuncular endocrine regulation, in particular the circadian rhythms that modulate the function of all organ systems. We outline the importance of cyclic endocrine regulation and the role that it may play in the development of MPS homunculi for the pharmacology, toxicology, and systems biology communities. Moreover, we discuss the critical end-organ hormone interactions that are most relevant for a typical coupled-OoC system, and the possible research applications of a missing endocrine system MicroFormulator (MES-µF) that could impose biological rhythms on in vitro models. By linking OoCs together through chemical messenger systems, advanced physiological phenomena relevant to pharmacokinetics and pharmacodynamics studies can be replicated. The concept of a MES-µF could be applied to other standard cell-culture systems such as well plates, thereby extending the concept of circadian hormonal regulation to much of in vitro biology. Impact statement Historically, cyclic endocrine modulation has been largely ignored within in vitro cell culture, in part because cultured cells typically have their media changed every day or two, precluding hourly adjustment of hormone concentrations to simulate circadian rhythms. As the Organ-on-Chip (OoC) community strives for greater physiological realism, the contribution of hormonal oscillations toward regulation of organ systems has been examined only in the context of reproductive organs, and circadian variation of the breadth of other hormones on most organs remains unaddressed. We illustrate the importance of cyclic endocrine modulation and the role that it plays within individual organ systems. The study of cyclic endocrine modulation within OoC systems will help advance OoC research to the point where it can reliably replicate in vitro key regulatory components of human physiology. This will help translate OoC work into pharmaceutical applications and connect the OoC community with the greater pharmacology and physiology communities.
Collapse
Affiliation(s)
- Kevin J. Cyr
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Systems Biology and Bioengineering Undergraduate Research Experience
| | - Omero M. Avaldi
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Systems Biology and Bioengineering Undergraduate Research Experience
| | - John P. Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education
- Department of Biomedical Engineering
- Department of Molecular Physiology and Biophysics
- Department of Physics and Astronomy, Vanderbilt University, Nashville TN, 37235, USA
| |
Collapse
|
49
|
Watson DE, Hunziker R, Wikswo JP. Fitting tissue chips and microphysiological systems into the grand scheme of medicine, biology, pharmacology, and toxicology. Exp Biol Med (Maywood) 2017; 242:1559-1572. [PMID: 29065799 DOI: 10.1177/1535370217732765] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microphysiological systems (MPS), which include engineered organoids (EOs), single organ/tissue chips (TCs), and multiple organs interconnected to create miniature in vitro models of human physiological systems, are rapidly becoming effective tools for drug development and the mechanistic understanding of tissue physiology and pathophysiology. The second MPS thematic issue of Experimental Biology and Medicine comprises 15 articles by scientists and engineers from the National Institutes of Health, the IQ Consortium, the Food and Drug Administration, and Environmental Protection Agency, an MPS company, and academia. Topics include the progress, challenges, and future of organs-on-chips, dissemination of TCs into Pharma, children's health protection, liver zonation, liver chips and their coupling to interconnected systems, gastrointestinal MPS, maturation of immature cardiomyocytes in a heart-on-a-chip, coculture of multiple cell types in a human skin construct, use of synthetic hydrogels to create EOs that form neural tissue models, the blood-brain barrier-on-a-chip, MPS models of coupled female reproductive organs, coupling MPS devices to create a body-on-a-chip, and the use of a microformulator to recapitulate endocrine circadian rhythms. While MPS hardware has been relatively stable since the last MPS thematic issue, there have been significant advances in cell sourcing, with increased reliance on human-induced pluripotent stem cells, and in characterization of the genetic and functional cell state in MPS bioreactors. There is growing appreciation of the need to minimize perfusate-to-cell-volume ratios and respect physiological scaling of coupled TCs. Questions asked by drug developers are followed by an analysis of the potential value, costs, and needs of Pharma. Of highest value and lowest switching costs may be the development of MPS disease models to aid in the discovery of disease mechanisms; novel compounds including probes, leads, and clinical candidates; and mechanism of action of drug candidates. Impact statement Microphysiological systems (MPS), which include engineered organoids and both individual and coupled organs-on-chips and tissue chips, are a rapidly growing topic of research that addresses the known limitations of conventional cellular monoculture on flat plastic - a well-perfected set of techniques that produces reliable, statistically significant results that may not adequately represent human biology and disease. As reviewed in this article and the others in this thematic issue, MPS research has made notable progress in the past three years in both cell sourcing and characterization. As the field matures, currently identified challenges are being addressed, and new ones are being recognized. Building upon investments by the Defense Advanced Research Projects Agency, National Institutes of Health, Food and Drug Administration, Defense Threat Reduction Agency, and Environmental Protection Agency of more than $200 million since 2012 and sizable corporate spending, academic and commercial players in the MPS community are demonstrating their ability to meet the translational challenges required to apply MPS technologies to accelerate drug development and advance toxicology.
Collapse
Affiliation(s)
| | - Rosemarie Hunziker
- 2 National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Wikswo
- 3 Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1807, USA
| |
Collapse
|
50
|
Schulze F, Gao X, Virzonis D, Damiati S, Schneider MR, Kodzius R. Air Quality Effects on Human Health and Approaches for Its Assessment through Microfluidic Chips. Genes (Basel) 2017; 8:E244. [PMID: 28953246 PMCID: PMC5664094 DOI: 10.3390/genes8100244] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/11/2017] [Accepted: 09/20/2017] [Indexed: 01/16/2023] Open
Abstract
Air quality depends on the various gases and particles present in it. Both natural phenomena and human activities affect the cleanliness of air. In the last decade, many countries experienced an unprecedented industrial growth, resulting in changing air quality values, and correspondingly, affecting our life quality. Air quality can be accessed by employing microchips that qualitatively and quantitatively determine the present gases and dust particles. The so-called particular matter 2.5 (PM2.5) values are of high importance, as such small particles can penetrate the human lung barrier and enter the blood system. There are cancer cases related to many air pollutants, and especially to PM2.5, contributing to exploding costs within the healthcare system. We focus on various current and potential future air pollutants, and propose solutions on how to protect our health against such dangerous substances. Recent developments in the Organ-on-Chip (OoC) technology can be used to study air pollution as well. OoC allows determination of pollutant toxicity and speeds up the development of novel pharmaceutical drugs.
Collapse
Affiliation(s)
- Frank Schulze
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), 10589 Berlin, Germany.
| | - Xinghua Gao
- iSmart, Materials Genome Institute, Shanghai University (SHU), Shanghai 201800, China.
| | - Darius Virzonis
- Department of Electrical Engineering, Kaunas University of Technology, 35212 Panevezys, Lithuania.
| | - Samar Damiati
- Department of Biochemistry, King Abdulaziz University, Jeddah 80203, Saudi Arabia.
- Institute for Synthetic Bioarchitecture, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria.
| | - Marlon R Schneider
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), 10589 Berlin, Germany.
| | - Rimantas Kodzius
- German Federal Institute for Risk Assessment (BfR), German Centre for the Protection of Laboratory Animals (Bf3R), 10589 Berlin, Germany.
- iSmart, Materials Genome Institute, Shanghai University (SHU), Shanghai 201800, China.
- Mathematics and Natural Sciences Department, The American University of Iraq, Sulaimani, Sulaymaniyah 46001, Iraq.
| |
Collapse
|