1
|
Barros BCSC, Barros DTL, Brock M, Toledo MS, Serrano SMT, Suzuki E, Ghaemmaghami AM. Secreted factors of Aspergillus fumigatus cause lung epithelial barrier disruption: A study using an air-liquid interface cell culture model. Med Mycol 2025; 63:myaf018. [PMID: 40036366 DOI: 10.1093/mmy/myaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 12/19/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
The effects of Aspergillus fumigatus-conditioned medium (AFCM) on the integrity of the Calu-3 cell lung epithelial barrier were investigated. AFCM led to a decrease in transepithelial electrical resistance and the disruption of the occludin network in the epithelial barrier. Preincubation with protease inhibitors reduced the effect of AFCM by ~ 90%, demonstrating the role of fungal proteases in epithelial barrier disruption. By mass spectrometry, we identified 494 unique proteins in AFCM, including 14 peptidases of different families. Together, these findings suggest that proteases secreted by A. fumigatus were able to modulate host epithelial barrier disruption in this fungal infection process.
Collapse
Affiliation(s)
- Bianca Carla Silva Campitelli Barros
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Debora Tereza Lucas Barros
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Matthias Brock
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| | - Marcos Sergio Toledo
- Departament of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Solange Maria Toledo Serrano
- Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo SP 05503-900, Brazil
| | - Erika Suzuki
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo SP 04023-062, Brazil
| | - Amir M Ghaemmaghami
- School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
2
|
Turner DL, Amoozadeh S, Baric H, Stanley E, Werder RB. Building a human lung from pluripotent stem cells to model respiratory viral infections. Respir Res 2024; 25:277. [PMID: 39010108 PMCID: PMC11251358 DOI: 10.1186/s12931-024-02912-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
To protect against the constant threat of inhaled pathogens, the lung is equipped with cellular defenders. In coordination with resident and recruited immune cells, this defence is initiated by the airway and alveolar epithelium following their infection with respiratory viruses. Further support for viral clearance and infection resolution is provided by adjacent endothelial and stromal cells. However, even with these defence mechanisms, respiratory viral infections are a significant global health concern, causing substantial morbidity, socioeconomic losses, and mortality, underlining the need to develop effective vaccines and antiviral medications. In turn, the identification of new treatment options for respiratory infections is critically dependent on the availability of tractable in vitro experimental models that faithfully recapitulate key aspects of lung physiology. For such models to be informative, it is important these models incorporate human-derived, physiologically relevant versions of all cell types that normally form part of the lungs anti-viral response. This review proposes a guideline using human induced pluripotent stem cells (iPSCs) to create all the disease-relevant cell types. iPSCs can be differentiated into lung epithelium, innate immune cells, endothelial cells, and fibroblasts at a large scale, recapitulating in vivo functions and providing genetic tractability. We advocate for building comprehensive iPSC-derived in vitro models of both proximal and distal lung regions to better understand and model respiratory infections, including interactions with chronic lung diseases.
Collapse
Affiliation(s)
- Declan L Turner
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Sahel Amoozadeh
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Hannah Baric
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Ed Stanley
- Murdoch Children's Research Institute, Melbourne, 3056, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia
| | - Rhiannon B Werder
- Murdoch Children's Research Institute, Melbourne, 3056, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, 3056, Australia.
- Novo Nordisk Foundation Centre for Stem Cell Medicine, reNEW Melbourne, Melbourne, 3056, Australia.
| |
Collapse
|
3
|
Nashihah AK, Muhammad Firdaus FI, Fauzi MB, Mobarak NN, Lokanathan Y. Role of Biomaterials in the Development of Epithelial Support in 3D In Vitro Airway Epithelium Development: A Systematic Review. Int J Mol Sci 2023; 24:14935. [PMID: 37834382 PMCID: PMC10573735 DOI: 10.3390/ijms241914935] [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: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
Respiratory diseases have a major impact on global health. The airway epithelium, which acts as a frontline defence, is one of the most common targets for inhaled allergens, irritants, or micro-organisms to enter the respiratory system. In the tissue engineering field, biomaterials play a crucial role. Due to the continuing high impact of respiratory diseases on society and the emergence of new respiratory viruses, in vitro airway epithelial models with high microphysiological similarities that are also easily adjustable to replicate disease models are urgently needed to better understand those diseases. Thus, the development of biomaterial scaffolds for the airway epithelium is important due to their function as a cell-support device in which cells are seeded in vitro and then are encouraged to lay down a matrix to form the foundations of a tissue for transplantation. Studies conducted in in vitro models are necessary because they accelerate the development of new treatments. Moreover, in comparatively controlled conditions, in vitro models allow for the stimulation of complex interactions between cells, scaffolds, and growth factors. Based on recent studies, the biomaterial scaffolds that have been tested in in vitro models appear to be viable options for repairing the airway epithelium and avoiding any complications. This review discusses the role of biomaterial scaffolds in in vitro airway epithelium models. The effects of scaffold, physicochemical, and mechanical properties in recent studies were also discussed.
Collapse
Affiliation(s)
- Ab Karim Nashihah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Fairuz Izan Muhammad Firdaus
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Mh. Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| | - Nadhratun Naiim Mobarak
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia UKM, Bangi 43600, Malaysia;
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia (F.I.M.F.); (M.B.F.)
| |
Collapse
|
4
|
Derman ID, Singh YP, Saini S, Nagamine M, Banerjee D, Ozbolat IT. Bioengineering and Clinical Translation of Human Lung and its Components. Adv Biol (Weinh) 2023; 7:e2200267. [PMID: 36658734 PMCID: PMC10121779 DOI: 10.1002/adbi.202200267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/18/2022] [Indexed: 01/21/2023]
Abstract
Clinical lung transplantation has rapidly established itself as the gold standard of treatment for end-stage lung diseases in a restricted group of patients since the first successful lung transplant occurred. Although significant progress has been made in lung transplantation, there are still numerous obstacles on the path to clinical success. The development of bioartificial lung grafts using patient-derived cells may serve as an alternative treatment modality; however, challenges include developing appropriate scaffold materials, advanced culture strategies for lung-specific multiple cell populations, and fully matured constructs to ensure increased transplant lifetime following implantation. This review highlights the development of tissue-engineered tracheal and lung equivalents over the past two decades, key problems in lung transplantation in a clinical environment, the advancements made in scaffolds, bioprinting technologies, bioreactors, organoids, and organ-on-a-chip technologies. The review aims to fill the lacuna in existing literature toward a holistic bioartificial lung tissue, including trachea, capillaries, airways, bifurcating bronchioles, lung disease models, and their clinical translation. Herein, the efforts are on bridging the application of lung tissue engineering methods in a clinical environment as it is thought that tissue engineering holds enormous promise for overcoming the challenges associated with the clinical translation of bioengineered human lung and its components.
Collapse
Affiliation(s)
- I. Deniz Derman
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Yogendra Pratap Singh
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Shweta Saini
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, India
| | - Momoka Nagamine
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
- Department of Chemistry, Penn State University; University Park, PA,16802, USA
| | - Dishary Banerjee
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University; University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University; University Park, PA, 16802, USA
- Biomedical Engineering Department, Penn State University; University Park, PA, 16802, USA
- Materials Research Institute, Penn State University; University Park, PA, 16802, USA
- Cancer Institute, Penn State University; University Park, PA, 16802, USA
- Neurosurgery Department, Penn State University; University Park, PA, 16802, USA
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| |
Collapse
|
5
|
Doryab A, Groll J. Biomimetic In Vitro Lung Models: Current Challenges and Future Perspective. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210519. [PMID: 36750972 DOI: 10.1002/adma.202210519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/10/2022] [Indexed: 06/18/2023]
Abstract
As post-COVID complications, chronic respiratory diseases are one of the foremost causes of mortality. The quest for a cure for this recent global challenge underlines that the lack of predictive in vitro lung models is one of the main bottlenecks in pulmonary preclinical drug development. Despite rigorous efforts to develop biomimetic in vitro lung models, the current cutting-edge models represent a compromise in numerous technological and biological aspects. Most advanced in vitro models are still in the "proof-of-concept" phase with a low clinical translation of the findings. On the other hand, advances in cellular and molecular studies are mainly based on relatively simple and unrealistic in vitro models. Herein, the current challenges and potential strategies toward not only bioinspired but truly biomimetic lung models are discussed.
Collapse
Affiliation(s)
- Ali Doryab
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Neuherberg, 85764, Munich, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), and Bavarian Polymer Institute (BPI), University of Würzburg, 97070, Würzburg, Germany
| |
Collapse
|
6
|
Optimization of Primary Human Bronchial Epithelial 3D Cell Culture with Donor-Matched Fibroblasts and Comparison of Two Different Culture Media. Int J Mol Sci 2023; 24:ijms24044113. [PMID: 36835529 PMCID: PMC9965758 DOI: 10.3390/ijms24044113] [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: 12/16/2022] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
In vitro airway models are increasingly important for pathomechanistic analyses of respiratory diseases. Existing models are limited in their validity by their incomplete cellular complexity. We therefore aimed to generate a more complex and meaningful three-dimensional (3D) airway model. Primary human bronchial epithelial cells (hbEC) were propagated in airway epithelial cell growth (AECG) or PneumaCult ExPlus medium. Generating 3D models, hbEC were airlifted and cultured on a collagen matrix with donor-matched bronchial fibroblasts for 21 days comparing two media (AECG or PneumaCult ALI (PC ALI)). 3D models were characterized by histology and immunofluorescence staining. The epithelial barrier function was quantified by transepithelial electrical resistance (TEER) measurements. The presence and function of ciliated epithelium were determined by Western blot and microscopy with high-speed camera. In 2D cultures, an increased number of cytokeratin 14-positive hbEC was present with AECG medium. In 3D models, AECG medium accounted for high proliferation, resulting in hypertrophic epithelium and fluctuating TEER values. Models cultured with PC ALI medium developed a functional ciliated epithelium with a stable epithelial barrier. Here, we established a 3D model with high in vivo-in vitro correlation, which has the potential to close the translational gap for investigations of the human respiratory epithelium in pharmacological, infectiological, and inflammatory research.
Collapse
|
7
|
Alternative lung cell model systems for toxicology testing strategies: Current knowledge and future outlook. Semin Cell Dev Biol 2023; 147:70-82. [PMID: 36599788 DOI: 10.1016/j.semcdb.2022.12.006] [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: 10/16/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023]
Abstract
Due to the current relevance of pulmonary toxicology (with focus upon air pollution and the inhalation of hazardous materials), it is important to further develop and implement physiologically relevant models of the entire respiratory tract. Lung model development has the aim to create human relevant systems that may replace animal use whilst balancing cost, laborious nature and regulatory ambition. There is an imperative need to move away from rodent models and implement models that mimic the holistic characteristics important in lung function. The purpose of this review is therefore, to describe and identify the various alternative models that are being applied towards assessing the pulmonary toxicology of inhaled substances, as well as the current and potential developments of various advanced models and how they may be applied towards toxicology testing strategies. These models aim to mimic various regions of the lung, as well as implementing different exposure methods with the addition of various physiologically relevent conditions (such as fluid-flow and dynamic movement). There is further progress in the type of models used with focus on the development of lung-on-a-chip technologies and bioprinting, as well as and the optimization of such models to fill current knowledge gaps within toxicology.
Collapse
|
8
|
Edwards VL, McComb E, Gleghorn JP, Forney L, Bavoil PM, Ravel J. Three-dimensional models of the cervicovaginal epithelia to study host-microbiome interactions and sexually transmitted infections. Pathog Dis 2022; 80:6655985. [PMID: 35927516 PMCID: PMC9419571 DOI: 10.1093/femspd/ftac026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/14/2022] [Accepted: 08/02/2022] [Indexed: 02/03/2023] Open
Abstract
2D cell culture systems have historically provided controlled, reproducible means to analyze host-pathogen interactions observed in the human reproductive tract. Although inexpensive, straightforward, and requiring a very short time commitment, these models recapitulate neither the functionality of multilayered cell types nor the associated microbiome that occurs in a human. Animal models have commonly been used to recreate the complexity of human infections. However, extensive modifications of animal models are required to recreate interactions that resemble those in the human reproductive tract. 3D cell culture models have emerged as alternative means of reproducing vital elements of human infections at a fraction of the cost of animal models and on a scale that allows for replicative experiments. Here, we describe a new 3D model that utilizes transwells with epithelial cells seeded apically and a basolateral extracellular matrix (ECM)-like layer. The model produced tissues with morphologic and physiological resemblance to human cervical and vaginal epithelia, including mucus levels produced by cervical cells. Infection by Chlamydia trachomatis and Neisseria gonorrhoeae was demonstrated, as well as the growth of bacterial species observed in the human vaginal microbiota. This enabled controlled mechanistic analyses of the interactions between host cells, the vaginal microbiota, and STI pathogens. Affordable and semi high-throughput 3D models of the cervicovaginal epithelia that are physiologically relevant by sustaining vaginal bacterial colonization, and facilitate studies of chlamydial and gonococcal infections.
Collapse
Affiliation(s)
- Vonetta L Edwards
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Larry Forney
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Patrik M Bavoil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Jacques Ravel
- Corresponding author: Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Health Science Research Facility (HSRDF), 670 W. Baltimore Street, Baltimore, MD 21201, United States. Tel: +1 410-706-5674; E-mail:
| |
Collapse
|
9
|
Doryab A, Schmid O. Bioactive Cell-Derived ECM Scaffold Forms a Unique Cellular Microenvironment for Lung Tissue Engineering. Biomedicines 2022; 10:biomedicines10081791. [PMID: 35892691 PMCID: PMC9394345 DOI: 10.3390/biomedicines10081791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases are one of the leading causes of death worldwide. Lung transplantation is currently the only causal therapeutic for lung diseases, which is restricted to end-stage disease and limited by low access to donor lungs. Lung tissue engineering (LTE) is a promising approach to regenerating a replacement for at least a part of the damaged lung tissue. Currently, lung regeneration is limited to a simplified local level (e.g., alveolar−capillary barrier) due to the sophisticated and complex structure and physiology of the lung. Here, we introduce an extracellular matrix (ECM)-integrated scaffold using a cellularization−decellularization−recellularization technique. This ECM-integrated scaffold was developed on our artificial co-polymeric BETA (biphasic elastic thin for air−liquid interface cell culture conditions) scaffold, which were initially populated with human lung fibroblasts (IMR90 cell line), as the main generator of ECM proteins. Due to the interconnected porous structure of the thin (<5 µm) BETA scaffold, the cells can grow on and infiltrate into the scaffold and deposit their own ECM. After a mild decellularization procedure, the ECM proteins remained on the scaffold, which now closely mimicked the cellular microenvironment of pulmonary cells more realistically than the plain artificial scaffolds. We assessed several decellularization methods and found that 20 mM NH4OH and 0.1% Triton X100 with subsequent DNase treatment completely removed the fibroblasts (from the first cellularization) and maintains collagen I and IV as the key ECM proteins on the scaffold. We also showed the repopulation of the primary fibroblast from human (without chronic lung disease (non-CLD) donors) and human bronchial epithelial (16HBE14o−) cells on the ECM-integrated BETA scaffold. With this technique, we developed a biomimetic scaffold that can mimic both the physico-mechanical properties and the native microenvironment of the lung ECM. The results indicate the potential of the presented bioactive scaffold for LTE application.
Collapse
|
10
|
Chmayssem A, Tanase CE, Verplanck N, Gougis M, Mourier V, Zebda A, Ghaemmaghami AM, Mailley P. New Microfluidic System for Electrochemical Impedance Spectroscopy Assessment of Cell Culture Performance: Design and Development of New Electrode Material. BIOSENSORS 2022; 12:bios12070452. [PMID: 35884254 PMCID: PMC9313146 DOI: 10.3390/bios12070452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 06/01/2023]
Abstract
Electrochemical impedance spectroscopy (EIS) is widely accepted as an effective and non-destructive method to assess cell health during cell-culture. However, there is a lack of compact devices compatible with microfluidic integration and microscopy that could provide the real-time and non-invasive monitoring of cell-cultures using EIS. In this paper, we reported the design and characterization of a modular EIS testing system based on a patented technology. This device was fabricated using easily processable methodologies including screen-printing of the impedance electrodes and molding or micromachining of the cell culture chamber with an easy assembly procedure. Accordingly, to obtain processable, biocompatible and sterilizable electrode materials that lower the impact of interfacial impedance on TEER (Transepithelial electrical resistance) measurements, and to enable concomitant microscopy observations, we optimized the formulation of the electrode inks and the design of the EIS electrodes, respectively. First, electrode materials were based on carbon biocompatible inks enriched with IrOx particles to obtain low interfacial impedance electrodes approaching the performances of classical non-biocompatible Ag/AgCl second-species electrodes. Secondly, we proposed three original electrode designs, which were compared to classical disk electrodes that were optically compatible with microscopy. We assessed the impact of the electrode design on the response of the impedance sensor using COMSOL Multiphysics. Finally, the performance of the impedance spectroscopy devices was assessed in vitro using human airway epithelial cell cultures.
Collapse
Affiliation(s)
- Ayman Chmayssem
- University Grenoble Alpes, CEA, LETI, DTBS, F-38000 Grenoble, France; (N.V.); (M.G.); (V.M.)
- University Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France;
| | - Constantin Edi Tanase
- Immunology & Immuno-Bioengineering Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (C.E.T.); (A.M.G.)
| | - Nicolas Verplanck
- University Grenoble Alpes, CEA, LETI, DTBS, F-38000 Grenoble, France; (N.V.); (M.G.); (V.M.)
| | - Maxime Gougis
- University Grenoble Alpes, CEA, LETI, DTBS, F-38000 Grenoble, France; (N.V.); (M.G.); (V.M.)
| | - Véronique Mourier
- University Grenoble Alpes, CEA, LETI, DTBS, F-38000 Grenoble, France; (N.V.); (M.G.); (V.M.)
| | - Abdelkader Zebda
- University Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France;
| | - Amir M. Ghaemmaghami
- Immunology & Immuno-Bioengineering Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (C.E.T.); (A.M.G.)
| | - Pascal Mailley
- University Grenoble Alpes, CEA, LETI, DTBS, F-38000 Grenoble, France; (N.V.); (M.G.); (V.M.)
| |
Collapse
|
11
|
Hammel JH, Zatorski JM, Cook SR, Pompano RR, Munson JM. Engineering in vitro immune-competent tissue models for testing and evaluation of therapeutics. Adv Drug Deliv Rev 2022; 182:114111. [PMID: 35031388 PMCID: PMC8908413 DOI: 10.1016/j.addr.2022.114111] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Advances in 3D cell culture, microscale fluidic control, and cellular analysis have enabled the development of more physiologically-relevant engineered models of human organs with precise control of the cellular microenvironment. Engineered models have been used successfully to answer fundamental biological questions and to screen therapeutics, but these often neglect key elements of the immune system. There are immune elements in every tissue that contribute to healthy and diseased states. Including immune function will be essential for effective preclinical testing of therapeutics for inflammatory and immune-modulated diseases. In this review, we first discuss the key components to consider in designing engineered immune-competent models in terms of physical, chemical, and biological cues. Next, we review recent applications of models of immunity for screening therapeutics for cancer, preclinical evaluation of engineered T cells, modeling autoimmunity, and screening vaccine efficacy. Future work is needed to further recapitulate immune responses in engineered models for the most informative therapeutic screening and evaluation.
Collapse
Affiliation(s)
- Jennifer H. Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Sophie R. Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA,Department of Biomedical Engineering, University of Virginia; Charlottesville, Virginia 22904, USA,Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| |
Collapse
|
12
|
Biological Models of the Lower Human Airways-Challenges and Special Requirements of Human 3D Barrier Models for Biomedical Research. Pharmaceutics 2021; 13:pharmaceutics13122115. [PMID: 34959396 PMCID: PMC8707984 DOI: 10.3390/pharmaceutics13122115] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 11/27/2022] Open
Abstract
In our review, we want to summarize the current status of the development of airway models and their application in biomedical research. We start with the very well characterized models composed of cell lines and end with the use of organoids. An important aspect is the function of the mucus as a component of the barrier, especially for infection research. Finally, we will explain the need for a nondestructive characterization of the barrier models using TEER measurements and live cell imaging. Here, organ-on-a-chip technology offers a great opportunity for the culture of complex airway models.
Collapse
|
13
|
Hammel JH, Cook SR, Belanger MC, Munson JM, Pompano RR. Modeling Immunity In Vitro: Slices, Chips, and Engineered Tissues. Annu Rev Biomed Eng 2021; 23:461-491. [PMID: 33872520 PMCID: PMC8277680 DOI: 10.1146/annurev-bioeng-082420-124920] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Modeling immunity in vitro has the potential to be a powerful tool for investigating fundamental biological questions, informing therapeutics and vaccines, and providing new insight into disease progression. There are two major elements to immunity that are necessary to model: primary immune tissues and peripheral tissues with immune components. Here, we systematically review progress made along three strategies to modeling immunity: ex vivo cultures, which preserve native tissue structure; microfluidic devices, which constitute a versatile approach to providing physiologically relevant fluid flow and environmental control; and engineered tissues, which provide precise control of the 3D microenvironment and biophysical cues. While many models focus on disease modeling, more primary immune tissue models are necessary to advance the field. Moving forward, we anticipate that the expansion of patient-specific models may inform why immunity varies from patient to patient and allow for the rapid comprehension and treatment of emerging diseases, such as coronavirus disease 2019.
Collapse
Affiliation(s)
- Jennifer H Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Sophie R Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Maura C Belanger
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Jennifer M Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA;
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA;
- Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903, USA
| |
Collapse
|
14
|
Soriano L, Khalid T, O’Brien FJ, O’Leary C, Cryan SA. A Tissue-Engineered Tracheobronchial In Vitro Co-Culture Model for Determining Epithelial Toxicological and Inflammatory Responses. Biomedicines 2021; 9:631. [PMID: 34199462 PMCID: PMC8226664 DOI: 10.3390/biomedicines9060631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/23/2021] [Accepted: 05/29/2021] [Indexed: 11/16/2022] Open
Abstract
Translation of novel inhalable therapies for respiratory diseases is hampered due to the lack of in vitro cell models that reflect the complexity of native tissue, resulting in many novel drugs and formulations failing to progress beyond preclinical assessments. The development of physiologically-representative tracheobronchial tissue analogues has the potential to improve the translation of new treatments by more accurately reflecting in vivo respiratory pharmacological and toxicological responses. Herein, advanced tissue-engineered collagen hyaluronic acid bilayered scaffolds (CHyA-B) previously developed within our group were used to evaluate bacterial and drug-induced toxicity and inflammation for the first time. Calu-3 bronchial epithelial cells and Wi38 lung fibroblasts were grown on either CHyA-B scaffolds (3D) or Transwell® inserts (2D) under air liquid interface (ALI) conditions. Toxicological and inflammatory responses from epithelial monocultures and co-cultures grown in 2D or 3D were compared, using lipopolysaccharide (LPS) and bleomycin challenges to induce bacterial and drug responses in vitro. The 3D in vitro model exhibited significant epithelial barrier formation that was maintained upon introduction of co-culture conditions. Barrier integrity showed differential recovery in CHyA-B and Transwell® epithelial cultures. Basolateral secretion of pro-inflammatory cytokines to bacterial challenge was found to be higher from cells grown in 3D compared to 2D. In addition, higher cytotoxicity and increased basolateral levels of cytokines were detected when epithelial cultures grown in 3D were challenged with bleomycin. CHyA-B scaffolds support the growth and differentiation of bronchial epithelial cells in a 3D co-culture model with different transepithelial resistance in comparison to the same co-cultures grown on Transwell® inserts. Epithelial cultures in an extracellular matrix like environment show distinct responses in cytokine release and metabolic activity compared to 2D polarised models, which better mimic in vivo response to toxic and inflammatory stimuli offering an innovative in vitro platform for respiratory drug development.
Collapse
Affiliation(s)
- Luis Soriano
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Tehreem Khalid
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Cian O’Leary
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland; (L.S.); (T.K.); (C.O.)
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- SFI Centre for Research in Medical Devices (CÚRAM), RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI University of Medicine and Health Sciences and Trinity College Dublin, D02 YN77 Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
15
|
Barron SL, Saez J, Owens RM. In Vitro Models for Studying Respiratory Host-Pathogen Interactions. Adv Biol (Weinh) 2021; 5:e2000624. [PMID: 33943040 PMCID: PMC8212094 DOI: 10.1002/adbi.202000624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Respiratory diseases and lower respiratory tract infections are among the leading cause of death worldwide and, especially given the recent severe acute respiratory syndrome coronavirus-2 pandemic, are of high and prevalent socio-economic importance. In vitro models, which accurately represent the lung microenvironment, are of increasing significance given the ethical concerns around animal work and the lack of translation to human disease, as well as the lengthy time to market and the attrition rates associated with clinical trials. This review gives an overview of the biological and immunological components involved in regulating the respiratory epithelium system in health, disease, and infection. The evolution from 2D to 3D cell biology and to more advanced technological integrated models for studying respiratory host-pathogen interactions are reviewed and provide a reference point for understanding the in vitro modeling requirements. Finally, the current limitations and future perspectives for advancing this field are presented.
Collapse
Affiliation(s)
- Sarah L. Barron
- Bioassay Impurities and QualityBiopharmaceuticals DevelopmentR&DAstraZenecaCambridgeCB21 6GPUK
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Janire Saez
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
16
|
3D Bioprinting for fabrication of tissue models of COVID-19 infection. Essays Biochem 2021; 65:503-518. [PMID: 34028514 DOI: 10.1042/ebc20200129] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Over the last few decades, the world has witnessed multiple viral pandemics, the current severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) pandemic being the worst and most devastating one, claiming millions of lives worldwide. Physicians, scientists, and engineers worldwide have joined hands in dealing with the current situation at an impressive speed and efficiency. One of the major reasons for the delay in response is our limited understanding of the mechanism of action and individual effects of the virus on different tissues and organs. Advances in 3D bioprinting have opened up a whole new area to explore and utilize the technology in fabricating models of these tissues and organs, recapitulating in vivo environment. These biomimetic models can not only be utilized in learning the infection pathways and drug toxicology studies but also minimize the need for animal models and shorten the time span for human clinical trials. The current review aims to integrate the existing developments in bioprinting techniques, and their implementation to develop tissue models, which has implications for SARS-CoV-2 infection. Future translation of these models has also been discussed with respect to the pandemic.
Collapse
|
17
|
Roy HS, Singh R, Ghosh D. SARS-CoV-2 and tissue damage: current insights and biomaterial-based therapeutic strategies. Biomater Sci 2021; 9:2804-2824. [PMID: 33666206 DOI: 10.1039/d0bm02077j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The effect of SARS-CoV-2 infection on humanity has gained worldwide attention and importance due to the rapid transmission, lack of treatment options and high mortality rate of the virus. While scientists across the world are searching for vaccines/drugs that can control the spread of the virus and/or reduce the risks associated with infection, patients infected with SARS-CoV-2 have been reported to have tissue/organ damage. With most tissues/organs having limited regenerative potential, interventions that prevent further damage or facilitate healing would be helpful. In the past few decades, biomaterials have gained prominence in the field of tissue engineering, in view of their major role in the regenerative process. Here we describe the effect of SARS-CoV-2 on multiple tissues/organs, and provide evidence for the positive role of biomaterials in aiding tissue repair. These findings are further extrapolated to explore their prospects as a therapeutic platform to address the tissue/organ damage that is frequently observed during this viral outbreak. This study suggests that the biomaterial-based approach could be an effective strategy for regenerating tissues/organs damaged by SARS-CoV-2.
Collapse
Affiliation(s)
- Himadri Shekhar Roy
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Rupali Singh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| | - Deepa Ghosh
- Department of Biological Science, Institute of Nanoscience and Technology (INST), Habitat Centre, Sector 64, Phase 10, Mohali-160062, Punjab, India.
| |
Collapse
|
18
|
Optimizations of In Vitro Mucus and Cell Culture Models to Better Predict In Vivo Gene Transfer in Pathological Lung Respiratory Airways: Cystic Fibrosis as an Example. Pharmaceutics 2020; 13:pharmaceutics13010047. [PMID: 33396283 PMCID: PMC7823756 DOI: 10.3390/pharmaceutics13010047] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 11/17/2022] Open
Abstract
The respiratory epithelium can be affected by many diseases that could be treated using aerosol gene therapy. Among these, cystic fibrosis (CF) is a lethal inherited disease characterized by airways complications, which determine the life expectancy and the effectiveness of aerosolized treatments. Beside evaluations performed under in vivo settings, cell culture models mimicking in vivo pathophysiological conditions can provide complementary insights into the potential of gene transfer strategies. Such models must consider multiple parameters, following the rationale that proper gene transfer evaluations depend on whether they are performed under experimental conditions close to pathophysiological settings. In addition, the mucus layer, which covers the epithelial cells, constitutes a physical barrier for gene delivery, especially in diseases such as CF. Artificial mucus models featuring physical and biological properties similar to CF mucus allow determining the ability of gene transfer systems to effectively reach the underlying epithelium. In this review, we describe mucus and cellular models relevant for CF aerosol gene therapy, with a particular emphasis on mucus rheology. We strongly believe that combining multiple pathophysiological features in single complex cell culture models could help bridge the gaps between in vitro and in vivo settings, as well as viral and non-viral gene delivery strategies.
Collapse
|
19
|
Shpichka A, Bikmulina P, Peshkova M, Kosheleva N, Zurina I, Zahmatkesh E, Khoshdel-Rad N, Lipina M, Golubeva E, Butnaru D, Svistunov A, Vosough M, Timashev P. Engineering a Model to Study Viral Infections: Bioprinting, Microfluidics, and Organoids to Defeat Coronavirus Disease 2019 (COVID-19). Int J Bioprint 2020; 6:302. [PMID: 33089000 PMCID: PMC7557357 DOI: 10.18063/ijb.v6i4.302] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
While the number of studies related to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) is constantly growing, it is essential to provide a framework of modeling viral infections. Therefore, this review aims to describe the background presented by earlier used models for viral studies and an approach to design an "ideal" tissue model for SARS-CoV-2 infection. Due to the previous successful achievements in antiviral research and tissue engineering, combining the emerging techniques such as bioprinting, microfluidics, and organoid formation are considered to be one of the best approaches to form in vitro tissue models. The fabrication of an integrated multi-tissue bioprinted platform tailored for SARS-CoV-2 infection can be a great breakthrough that can help defeat coronavirus disease in 2019.
Collapse
Affiliation(s)
- Anastasia Shpichka
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Polina Bikmulina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Maria Peshkova
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Nastasia Kosheleva
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
- Department of Embryology, Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Irina Zurina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Ensieh Zahmatkesh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marina Lipina
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, Moscow, Russia
| | - Elena Golubeva
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Denis Butnaru
- Rector’s Office, Sechenov University, Moscow, Russia
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
- Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia
| |
Collapse
|
20
|
O'Leary C, Soriano L, Fagan-Murphy A, Ivankovic I, Cavanagh B, O'Brien FJ, Cryan SA. The Fabrication and in vitro Evaluation of Retinoic Acid-Loaded Electrospun Composite Biomaterials for Tracheal Tissue Regeneration. Front Bioeng Biotechnol 2020; 8:190. [PMID: 32266229 PMCID: PMC7103641 DOI: 10.3389/fbioe.2020.00190] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/27/2020] [Indexed: 12/24/2022] Open
Abstract
Although relatively rare, major trauma to the tracheal region of the airways poses a significant clinical challenge with few effective treatments. Bioengineering and regenerative medicine strategies have the potential to create biocompatible, implantable biomaterial scaffolds, with the capacity to restore lost tissue with functional neo-trachea. The main goal of this study was to develop a nanofibrous polycaprolactone-chitosan (PCL-Chitosan) scaffold loaded with a signaling molecule, all-trans retinoic acid (atRA), as a novel biomaterial approach for tracheal tissue engineering. Using the Spraybase® electrospinning platform, polymer concentration, solvent selection, and instrument parameters were optimized to yield a co-polymer with nanofibers of 181-197 nm in diameter that mimicked tracheobronchial tissue architecture. Thereafter, scaffolds were assessed for their biocompatibility and capacity to induce mucociliary functionalization using the Calu-3 cell line. PCL-Chitosan scaffolds were found to be biocompatible in nature and support Calu-3 cell viability over a 14 day time period. Additionally, the inclusion of atRA did not compromise Calu-3 cell viability, while still achieving an efficient encapsulation of the signaling molecule over a range of atRA concentrations. atRA release from scaffolds led to an increase in mucociliary gene expression at high scaffold loading doses, with augmented MUC5AC and FOXJ1 detected by RT-PCR. Overall, this scaffold integrates a synthetic polymer that has been used in human tracheal stents, a natural polymer generally regarded as safe (GRAS), and a drug with decades of use in patients. Coupled with the scalable nature of electrospinning as a fabrication method, all of these characteristics make the biomaterial outlined in this study amenable as an implantable device for an unmet clinical need in tracheal replacement.
Collapse
Affiliation(s)
- Cian O'Leary
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Luis Soriano
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aidan Fagan-Murphy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ivana Ivankovic
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Center for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- SFI Center for Research in Medical Devices (CÚRAM), Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Center for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
21
|
Kumar P, Ciftci S, Barthes J, Knopf‐Marques H, Muller CB, Debry C, Vrana NE, Ghaemmaghami AM. A composite Gelatin/hyaluronic acid hydrogel as an ECM mimic for developing mesenchymal stem cell‐derived epithelial tissue patches. J Tissue Eng Regen Med 2019; 14:45-57. [DOI: 10.1002/term.2962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/29/2019] [Accepted: 09/04/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Pramod Kumar
- Immunology and Tissue Modelling Group, School of Life Science, Faculty of Medicine and Health SciencesUniversity of Nottingham Nottingham UK
| | - Sait Ciftci
- INSERM UMR 1121 Strasbourg France
- Service Oto‐Rhino‐LaryngologieHôpitaux Universitaires de Strasbourg Strasbourg France
| | - Julien Barthes
- INSERM UMR 1121 Strasbourg France
- Protip Medical Strasbourg France
| | - Helena Knopf‐Marques
- INSERM UMR 1121 Strasbourg France
- Faculté de Chirurgie DentaireUniversité de Strasbourg Strasbourg France
| | | | - Christian Debry
- INSERM UMR 1121 Strasbourg France
- Service Oto‐Rhino‐LaryngologieHôpitaux Universitaires de Strasbourg Strasbourg France
| | - Nihal E. Vrana
- INSERM UMR 1121 Strasbourg France
- Protip Medical Strasbourg France
| | - Amir M. Ghaemmaghami
- Immunology and Tissue Modelling Group, School of Life Science, Faculty of Medicine and Health SciencesUniversity of Nottingham Nottingham UK
| |
Collapse
|
22
|
Lu Q, El-Hashash AHK. Cell-based therapy for idiopathic pulmonary fibrosis. Stem Cell Investig 2019; 6:22. [PMID: 31559309 PMCID: PMC6737434 DOI: 10.21037/sci.2019.06.09] [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: 04/13/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an example of interstitial lung diseases that is characterized by chronic, progressive, and fibrotic lung injuries. During lung fibrosis, normal healthy lung tissues are replaced by remarkably destroyed alveolar architecture and altered extracellular cell matrix. These changes eventually cause severe disruption of the tightly-controlled gas exchange process and reduction of lung compliance that ultimately lead to both respiratory failure and death. In the last decade, progress has been made toward understanding the pathogenesis of pulmonary fibrosis, and two novel disease-modifying therapies were approved. However, finding more effective treatments for pulmonary fibrosis is still a challenge, with its incidence continues to increase globally, which is associated with significantly high mortality, morbidity and economical healthcare burden. Different stem cell types have recently emerged as a promising therapy for human diseases, including lung fibrosis, with numerous studies on the identification, characterization, proliferation and differentiation of stem cells. A large body of both basic and pre-clinical research on stem cells has been recently translated to patient care worldwide. Herein, we review recent advances in our understanding of the pathophysiology of IPF, and types of cells used in IPF cell-based therapies, including alveolar and mixed lung epithelial cells, different stem cell types (MSCs, ADSCs, IPSCs…etc.), endogenous lung tissue-specific stem cells, and circulating endothelial progenitors (EPCs). We also discuss recent studies on the applications of these cells in IPF therapy and their delivery routes, effective doses for cell therapy, and timing of delivery. Finally, we discuss attractive recent and current clinical trials conducted on cell-based therapy for IPF.
Collapse
Affiliation(s)
- Qi Lu
- The University of Edinburgh-Zhejiang International campus (UoE-ZJU Institute), Haining, China
- Centre of Stem Cell and Regenerative Medicine Schools of Medicine & Basic Medicine, Hangzhou, China
| | - Ahmed H. K. El-Hashash
- The University of Edinburgh-Zhejiang International campus (UoE-ZJU Institute), Haining, China
- Centre of Stem Cell and Regenerative Medicine Schools of Medicine & Basic Medicine, Hangzhou, China
| |
Collapse
|
23
|
Derakhshani S, Kurz A, Japtok L, Schumacher F, Pilgram L, Steinke M, Kleuser B, Sauer M, Schneider-Schaulies S, Avota E. Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium. Front Immunol 2019; 10:1294. [PMID: 31231395 PMCID: PMC6560165 DOI: 10.3389/fimmu.2019.01294] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/21/2019] [Indexed: 12/16/2022] Open
Abstract
Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit.
Collapse
Affiliation(s)
| | - Andreas Kurz
- Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Lukasz Japtok
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Lisa Pilgram
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Maria Steinke
- Fraunhofer Institute for Silicate Research ISC, Chair of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Markus Sauer
- Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
24
|
Patient JD, Hajiali H, Harris K, Abrahamsson B, Tannergren C, White LJ, Ghaemmaghami AM, Williams PM, Roberts CJ, Rose FRAJ. Nanofibrous Scaffolds Support a 3D in vitro Permeability Model of the Human Intestinal Epithelium. Front Pharmacol 2019; 10:456. [PMID: 31133850 PMCID: PMC6524416 DOI: 10.3389/fphar.2019.00456] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
Advances in drug research not only depend on high throughput screening to evaluate large numbers of lead compounds but also on the development of in vitro models which can simulate human tissues in terms of drug permeability and functions. Potential failures, such as poor permeability or interaction with efflux drug transporters, can be identified in epithelial Caco-2 monolayer models and can impact a drug candidate's progression onto the next stages of the drug development process. Whilst monolayer models demonstrate reasonably good prediction of in vivo permeability for some compounds, more developed in vitro tools are needed to assess new entities that enable closer in vivo in vitro correlation. In this study, an in vitro model of the human intestinal epithelium was developed by utilizing nanofibers, fabricated using electrospinning, to mimic the structure of the basement membrane. We assessed Caco-2 cell response to these materials and investigated the physiological properties of these cells cultured on the fibrous supports, focusing on barrier integrity and drug-permeability properties. The obtained data illustrate that 2D Caco-2 Transwell® cultures exhibit artificially high trans-epithelial electrical resistance (TEER) compared to cells cultured on the 3D nanofibrous scaffolds which show TEER values similar to ex vivo porcine tissue (also measured in this study). Furthermore, our results demonstrate that the 3D nanofibrous scaffolds influence the barrier integrity of the Caco-2 monolayer to confer drug-absorption properties that more closely mimic native gut tissue particularly for studying passive epithelial transport. We propose that this 3D model is a suitable in vitro model for investigating drug absorption and intestinal metabolism.
Collapse
Affiliation(s)
- Jamie D. Patient
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Hadi Hajiali
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | | | | | | | - Lisa J. White
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Amir M. Ghaemmaghami
- School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Philip M. Williams
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Clive J. Roberts
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | | |
Collapse
|
25
|
Ahmed S, Chauhan VM, Ghaemmaghami AM, Aylott JW. New generation of bioreactors that advance extracellular matrix modelling and tissue engineering. Biotechnol Lett 2019; 41:1-25. [PMID: 30368691 PMCID: PMC6313369 DOI: 10.1007/s10529-018-2611-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
Bioreactors hold a lot of promise for tissue engineering and regenerative medicine applications. They have multiple uses including cell cultivation for therapeutic production and for in vitro organ modelling to provide a more physiologically relevant environment for cultures compared to conventional static conditions. Bioreactors are often used in combination with scaffolds as the nutrient flow can enhance oxygen and diffusion throughout the 3D constructs to prevent the formation of necrotic cores. A variety of scaffolds have been fabricated to achieve a structural architecture that mimic native extracellular matrix. Future developments of in vitro models will incorporate the ability to non-invasively monitor the cellular microenvironment to enhance the understanding of in vitro conditions. This review details current advancements in bioreactor and scaffold systems and provides insight on how in vitro models can be augmented for future biomedical applications.
Collapse
Affiliation(s)
- Shehnaz Ahmed
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| | - Veeren M. Chauhan
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| | - Amir M. Ghaemmaghami
- School of Life Sciences, University of Nottingham, Life Sciences Building, University Park, Nottingham, NG7 2RD UK
| | - Jonathan W. Aylott
- School of Pharmacy, University of Nottingham, Boots Sciences Building, University Park, Nottingham, UK
| |
Collapse
|
26
|
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
|
27
|
Castellani S, Di Gioia S, di Toma L, Conese M. Human Cellular Models for the Investigation of Lung Inflammation and Mucus Production in Cystic Fibrosis. Anal Cell Pathol (Amst) 2018; 2018:3839803. [PMID: 30581723 PMCID: PMC6276497 DOI: 10.1155/2018/3839803] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/04/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation, oxidative stress, mucus plugging, airway remodeling, and respiratory infections are the hallmarks of the cystic fibrosis (CF) lung disease. The airway epithelium is central in the innate immune responses to pathogens colonizing the airways, since it is involved in mucociliary clearance, senses the presence of pathogens, elicits an inflammatory response, orchestrates adaptive immunity, and activates mesenchymal cells. In this review, we focus on cellular models of the human CF airway epithelium that have been used for studying mucus production, inflammatory response, and airway remodeling, with particular reference to two- and three-dimensional cultures that better recapitulate the native airway epithelium. Cocultures of airway epithelial cells, macrophages, dendritic cells, and fibroblasts are instrumental in disease modeling, drug discovery, and identification of novel therapeutic targets. Nevertheless, they have to be implemented in the CF field yet. Finally, novel systems hijacking on tissue engineering, including three-dimensional cocultures, decellularized lungs, microfluidic devices, and lung organoids formed in bioreactors, will lead the generation of relevant human preclinical respiratory models a step forward.
Collapse
Affiliation(s)
- Stefano Castellani
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lorena di Toma
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
28
|
Jung JP, Lin WH, Riddle MJ, Tolar J, Ogle BM. A 3D in vitro model of the dermoepidermal junction amenable to mechanical testing. J Biomed Mater Res A 2018; 106:3231-3238. [PMID: 30208260 PMCID: PMC6283247 DOI: 10.1002/jbm.a.36519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/20/2018] [Accepted: 07/31/2018] [Indexed: 01/06/2023]
Abstract
Recessive dystrophic Epidermolysis Bullosa (RDEB) is caused by mutations in collagen‐type VII gene critical for the dermoepidermal junction (DEJ) formation. Neither tissues of animal models nor currently available in vitro models are amenable to the quantitative assessment of mechanical adhesion between dermal and epidermal layers. Here, we created a 3D in vitro DEJ model using extracellular matrix (ECM) proteins of the DEJ anchored to a poly(ethylene glycol)‐based slab (termed ECM composites) and seeded with human keratinocytes and dermal fibroblasts. Keratinocytes and fibroblasts of healthy individuals were well maintained in the ECM composite and showed the expression of collagen type VII over a 2‐week period. The ECM composites with healthy keratinocytes and fibroblasts exhibited yield stress associated with the separation of the model DEJ at 0.268 ± 0.057 kPa. When we benchmarked this measure of adhesive strength with that of the model DEJ fabricated with cells of individuals with RDEB, the yield stress was significantly lower (0.153 ± 0.064 kPa) consistent with our current mechanistic understanding of RDEB. In summary, a 3D in vitro model DEJ was developed for quantification of mechanical adhesion between epidermal‐ and dermal‐mimicking layers, which can be utilized for assessment of mechanical adhesion of the model DEJ applicable for Epidermolysis Bullosa‐associated therapeutics. © 2018 The Authors. Journal Of Biomedical Materials Research Part A Published By Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3231–3238, 2018.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana
| | - Wei-Han Lin
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Megan J Riddle
- Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
29
|
Svensson M, Chen P. Human Organotypic Respiratory Models. Curr Top Microbiol Immunol 2018:29-54. [PMID: 29808337 DOI: 10.1007/82_2018_91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Biomedical research aiming to understand the molecular basis of human lung tissue development, homeostasis and disease, or to develop new therapies for human respiratory diseases, requires models that faithfully recapitulate the human condition. This has stimulated biologists and engineers to develop in vitro organotypic models mimicking human respiratory tissues. In this chapter, we provide examples of different types of model systems ranging from simple unicellular cultures to more complex multicellular systems. The models contain, in varying degree, cell types present in real tissue in combination with different extracellular matrix components that can critically affect cell phenotype and function. We also describe how organotypic respiratory models can be combined with human innate immune cells, to better recapitulate tissue inflammation, a key component in, for example, infectious diseases. These models have the potential to provide new insights into lung physiology, tissue infection and inflammation, disease mechanisms, as well as provide a platform for identification of novel targets and screening of candidate drugs in human lung disorders.
Collapse
Affiliation(s)
- Mattias Svensson
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden.
| | - Puran Chen
- F59, Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86, Stockholm, Sweden
| |
Collapse
|
30
|
De Rudder C, Calatayud Arroyo M, Lebeer S, Van de Wiele T. Modelling upper respiratory tract diseases: getting grips on host-microbe interactions in chronic rhinosinusitis using in vitro technologies. MICROBIOME 2018; 6:75. [PMID: 29690931 PMCID: PMC5913889 DOI: 10.1186/s40168-018-0462-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 04/17/2018] [Indexed: 05/27/2023]
Abstract
Chronic rhinosinusitis (CRS) is a chronic inflammation of the mucosa of the nose and paranasal sinuses affecting approximately 11% of the adult population in Europe. Inadequate immune responses, as well as a dysbiosis of the sinonasal microbiota, have been put forward as aetiological factors of the disease. However, despite the prevalence of this disease, there is no consensus on the aetiology and mechanisms of pathogenesis of CRS. Further research requires in vitro models mimicking the healthy and diseased host environment along with the sinonasal microbiota. This review aims to provide an overview of CRS model systems and proposes in vitro modelling strategies to conduct mechanistic research in an ecological framework on the sinonasal microbiota and its interactions with the host in health and CRS.
Collapse
Affiliation(s)
- Charlotte De Rudder
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Marta Calatayud Arroyo
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Sarah Lebeer
- Research Group of Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium.
| |
Collapse
|
31
|
Zscheppang K, Berg J, Hedtrich S, Verheyen L, Wagner DE, Suttorp N, Hippenstiel S, Hocke AC. Human Pulmonary 3D Models For Translational Research. Biotechnol J 2018; 13:1700341. [PMID: 28865134 PMCID: PMC7161817 DOI: 10.1002/biot.201700341] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/23/2017] [Indexed: 12/13/2022]
Abstract
Lung diseases belong to the major causes of death worldwide. Recent innovative methodological developments now allow more and more for the use of primary human tissue and cells to model such diseases. In this regard, the review covers bronchial air-liquid interface cultures, precision cut lung slices as well as ex vivo cultures of explanted peripheral lung tissue and de-/re-cellularization models. Diseases such as asthma or infections are discussed and an outlook on further areas for development is given. Overall, the progress in ex vivo modeling by using primary human material could make translational research activities more efficient by simultaneously fostering the mechanistic understanding of human lung diseases while reducing animal usage in biomedical research.
Collapse
Affiliation(s)
- Katja Zscheppang
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Johanna Berg
- Department of BiotechnologyTechnical University of BerlinGustav‐Meyer‐Allee 25Berlin 13335Germany
| | - Sarah Hedtrich
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Leonie Verheyen
- Institute for PharmacyPharmacology and ToxicologyFreie Universität BerlinBerlinGermany
| | - Darcy E. Wagner
- Helmholtz Zentrum Munich, Lung Repair and Regeneration Unit, Comprehensive Pneumology CenterMember of the German Center for Lung ResearchMunichGermany
| | - Norbert Suttorp
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Stefan Hippenstiel
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| | - Andreas C. Hocke
- Dept. of Internal Medicine/Infectious and Respiratory DiseasesCharité − Universitätsmedizin BerlinCharitèplatz 1Berlin 10117Germany
| |
Collapse
|
32
|
Novel Models to Study Stromal Cell-Leukocyte Interactions in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1060:131-146. [PMID: 30155626 DOI: 10.1007/978-3-319-78127-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To study human immunology in general and stromal immunology in particular, it is highly motivated to move from monolayers to 3D cultures, such as organotypic models, that better mimic the function of living tissue. These models can potentially contain most if not all cell types present in tissues, in combination with different extracellular matrix components that can critically affect cell phenotype. Besides their well-established use in studies of tissue-specific cells, such as epithelial cells, endothelial cells and stromal fibroblasts in combination with extracellular components, these models have also been shown to be valuable to study how tissue participates in the regulation of leukocyte differentiation and function. Organotypic models with leukocytes represent novel powerful tools to study human stromal immunology and mechanisms involved in the regulation of leukocyte functions and inflammatory processes in human health and disease. In particular, these models are robust, long-lived and reproducible and allow monitoring of disease progression in real time, as well as the mixing of cellular constituents from healthy and pathological tissues. These models are also easy to manipulate, either genetically or by adding external stimulants, such as cytokines and pathogens, to mimic pathological conditions. It is thus not surprising that these models are proposed to be useful in toxicology screening assays, evaluating therapeutic efficacy of drugs and antibiotics, as well as in personalized medicine. Within this chapter, the most recent developments in creating organotypic models for the purpose of study of human leukocyte and stromal cell interactions, in health and disease, will be discussed, in particular focusing on live imaging. Special emphasis will be given on an organotypic model resembling human lung and its usefulness in studying the fine control of physiological and pathological processes in human health and disease. Using these models in studies on human stromal cell and leukocyte interactions will likely help identifying novel disease traits and may point out new potential targets to monitor and treat human diseases.
Collapse
|
33
|
Cha BH, Shin SR, Leijten J, Li YC, Singh S, Liu JC, Annabi N, Abdi R, Dokmeci MR, Vrana NE, Ghaemmaghami AM, Khademhosseini A. Integrin-Mediated Interactions Control Macrophage Polarization in 3D Hydrogels. Adv Healthc Mater 2017; 6:10.1002/adhm.201700289. [PMID: 28782184 PMCID: PMC5677560 DOI: 10.1002/adhm.201700289] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/17/2017] [Indexed: 12/23/2022]
Abstract
Adverse immune reactions prevent clinical translation of numerous implantable devices and materials. Although inflammation is an essential part of tissue regeneration, chronic inflammation ultimately leads to implant failure. In particular, macrophage polarity steers the microenvironment toward inflammation or wound healing via the induction of M1 and M2 macrophages, respectively. Here, this paper demonstrates that macrophage polarity within biomaterials can be controlled through integrin-mediated interactions between human monocytic THP-1 cells and collagen-derived matrix. Surface marker, gene expression, biochemical, and cytokine profiling consistently indicate that THP-1 cells within a biomaterial lacking cell attachment motifs yield proinflammatory M1 macrophages, whereas biomaterials with attachment sites in the presence of interleukin-4 (IL-4) induce an anti-inflammatory M2-like phenotype and propagate the effect of IL-4 in induction of M2-like macrophages. Importantly, integrin α2β1 plays a pivotal role as its inhibition blocks the induction of M2 macrophages. The influence of the microenvironment of the biomaterial over macrophage polarity is further confirmed by its ability to modulate the effect of IL-4 and lipopolysaccharide, which are potent inducers of M2 or M1 phenotypes, respectively. Thus, this study represents a novel, versatile, and effective strategy to steer macrophage polarity through integrin-mediated 3D microenvironment for biomaterial-based programming.
Collapse
Affiliation(s)
- Byung-Hyun Cha
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Su Ryon Shin
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, 7500, AE, Enschede, The Netherlands
| | - Yi-Chen Li
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Sonali Singh
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Julie C Liu
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Davidson School of Chemical Engineering and Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Nasim Annabi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Reza Abdi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Transplant Research Center, Renal Division, Brigham and Women's Hospital and Children's Hospital, Boston, MA, 02115, USA
| | - Mehmet R Dokmeci
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Nihal Engin Vrana
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Fundamental Research Unit, Protip Medical, 8 Place de l'Hôpital, 67000, Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S 1121, "Biomatériaux et Bioingénierie", 11 rue Humann, 67085, Strasbourg Cedex, France
| | - Amir M Ghaemmaghami
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
- Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul, 143-701, Republic of Korea
- Nanotechnology Center, King Abdulaziz University, Jeddah, 21569, Saudi Arabia
| |
Collapse
|
34
|
Mizoguchi I, Ohashi M, Chiba Y, Hasegawa H, Xu M, Owaki T, Yoshimoto T. Prediction of Chemical Respiratory and Contact Sensitizers by OX40L Expression in Dendritic Cells Using a Novel 3D Coculture System. Front Immunol 2017; 8:929. [PMID: 28824649 PMCID: PMC5543289 DOI: 10.3389/fimmu.2017.00929] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
The use of animal models in chemical safety testing will be significantly limited due to the recent introduction of the 3Rs principle of animal experimentation in research. Although several in vitro assays to predict the sensitizing potential of chemicals have been developed, these methods cannot distinguish chemical respiratory sensitizers and skin sensitizers. In the present study, we describe a novel in vitro assay that can discriminate respiratory sensitizers from chemical skin sensitizers by taking advantage of the fundamental difference between their modes of action, namely the development of the T helper 2 immune response, which is critically important for respiratory sensitization. First, we established a novel three-dimensional (3D) coculture system of human upper airway epithelium using a commercially available scaffold. It consists of human airway epithelial cell line BEAS-2B, immature dendritic cells (DCs) derived from human peripheral blood CD14+ monocytes, and human lung fibroblast cell line MRC-5. Respective cells were first cultured in individual scaffolds and subsequently assembled into a 3D multi-cell tissue model to more closely mimic the in vivo situation. Then, three typical chemicals that are known respiratory sensitizers (ortho-phthaldialdehyde, hexamethylene diisocyanate, and trimellitic anhydride) and skin sensitizers (oxazolone, formaldehyde, and dinitrochlorobenzene) were added individually to the 3D coculture system. Immunohistochemical analysis revealed that DCs do not migrate into other scaffolds under the experimental conditions. Therefore, the 3D structure was disassembled and real-time reverse transcriptase-PCR analysis was performed in individual scaffolds to analyze the expression levels of molecules critical for Th2 differentiation such as OX40 ligand (OX40L), interleukin (IL)-4, IL-10, IL-33, and thymic stromal lymphopoietin. Both sensitizers showed similarly augmented expression of DC maturation markers (e.g., CD86), but among these molecules, OX40L expression in DCs was most consistently and significantly enhanced by respiratory sensitizers as compared to that by skin sensitizers. Thus, we have established a 3D coculture system mimicking the airway upper epithelium that may be successfully applied to discriminate chemical respiratory sensitizers from skin sensitizers by measuring the critical molecule for Th2 differentiation, OX40L, in DCs.
Collapse
Affiliation(s)
- Izuru Mizoguchi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Mio Ohashi
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yukino Chiba
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hideaki Hasegawa
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Mingli Xu
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Toshiyuki Owaki
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Takayuki Yoshimoto
- Department of Immunoregulation, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
35
|
Abstract
Neisseria meningitidis is a harmless commensal bacterium finely adapted to humans. Unfortunately, under “privileged” conditions, it adopts a “devious” lifestyle leading to uncontrolled behavior characterized by the unleashing of molecular weapons causing potentially lethal disease such as sepsis and acute meningitis. Indeed, despite the lack of a classic repertoire of virulence genes in
N. meningitidis separating commensal from invasive strains, molecular epidemiology and functional genomics studies suggest that carriage and invasive strains belong to genetically distinct populations characterized by an exclusive pathogenic potential. In the last few years, “omics” technologies have helped scientists to unwrap the framework drawn by
N. meningitidis during different stages of colonization and disease. However, this scenario is still incomplete and would benefit from the implementation of physiological tissue models for the reproduction of mucosal and systemic interactions
in vitro. These emerging technologies supported by recent advances in the world of stem cell biology hold the promise for a further understanding of
N. meningitidis pathogenesis.
Collapse
|
36
|
Rostam HM, Reynolds PM, Alexander MR, Gadegaard N, Ghaemmaghami AM. Image based Machine Learning for identification of macrophage subsets. Sci Rep 2017; 7:3521. [PMID: 28615717 PMCID: PMC5471192 DOI: 10.1038/s41598-017-03780-z] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/03/2017] [Indexed: 11/29/2022] Open
Abstract
Macrophages play a crucial rule in orchestrating immune responses against pathogens and foreign materials. Macrophages have remarkable plasticity in response to environmental cues and are able to acquire a spectrum of activation status, best exemplified by pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes at the two ends of the spectrum. Characterisation of M1 and M2 subsets is usually carried out by quantification of multiple cell surface markers, transcription factors and cytokine profiles. These approaches are time-consuming, require large numbers of cells and are resource intensive. In this study, we used machine learning algorithms to develop a simple and fast imaging-based approach that enables automated identification of different macrophage functional phenotypes using their cell size and morphology. Fluorescent microscopy was used to assess cell morphology of different cell types which were stained for nucleus and actin distribution using DAPI and phalloidin respectively. By only analysing their morphology we were able to identify M1 and M2 phenotypes effectively and could distinguish them from naïve macrophages and monocytes with an average accuracy of 90%. Thus we suggest high-content and automated image analysis can be used for fast phenotyping of functionally diverse cell populations with reasonable accuracy and without the need for using multiple markers.
Collapse
Affiliation(s)
- Hassan M Rostam
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.,Department of Biology, University of Garmian, Kalar, Kurdistan, Iraq
| | - Paul M Reynolds
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK
| | - Morgan R Alexander
- Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow, G12 8LT, UK.
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
37
|
Rimington TL, Hodge E, Billington CK, Bhaker S, K C B, Kilty I, Jelinsky S, Hall IP, Sayers I. Defining the inflammatory signature of human lung explant tissue in the presence and absence of glucocorticoid. F1000Res 2017; 6:460. [PMID: 28721202 PMCID: PMC5497818 DOI: 10.12688/f1000research.10961.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/10/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Airway inflammation is a feature of many respiratory diseases and there is a need for newer, more effective anti-inflammatory compounds. The aim of this study was to develop an
ex vivo human lung explant model which can be used to help study the mechanisms underlying inflammatory responses and which can provide a tool to aid drug discovery for inflammatory respiratory diseases such as asthma and COPD. Method: Parenchymal lung tissue from 6 individual donors was dissected and cultured with two pro-inflammatory stimuli, lipopolysaccharide (LPS) (1 µg/ml) and interleukin-1 beta (IL-1β) (10 ng/ml) in the presence or absence of dexamethasone (1 µM). Inflammatory responses were assessed using Luminex analysis of tissue culture supernatants to measure levels of 21 chemokines, growth factors and cytokines. Results: A robust and reproducible inflammatory signal was detected across all donors for 12 of the analytes measured following LPS stimulation with a modest fold increase (<2-fold) in levels of CCL22, IL-4, and IL-2; increases of 2-4-fold in levels of CXCL8, VEGF and IL-6 and increases >4-fold in CCL3, CCL4, GM-CSF, IL-10, TNF-α and IL-1β. The inflammatory signal induced by IL-1β stimulation was less than that observed with LPS but resulted in elevated levels of 7 analytes (CXCL8, CCL3, CCL4, GM-CSF, IL-6, IL-10 and TNF-α). The inflammatory responses induced by both stimulations was supressed by dexamethasone for the majority of analytes. Conclusions: These data provide proof of concept that this
ex vivo human lung explant model is responsive to inflammatory signals and could be used to investigate the anti-inflammatory effects of existing and novel compounds. In addition this model could be used to help define the mechanisms and pathways involved in development of inflammatory airway disease. Abbreviations: COPD: Chronic Obstructive Pulmonary Disease; ICS: inhaled corticosteroids; LPS: lipopolysaccharide; IL-1β: interleukin-1 beta; PSF: penicillin, streptomycin and fungizone
Collapse
Affiliation(s)
- Tracy L Rimington
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | | | - Sangita Bhaker
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Binaya K C
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK.,Department of Mechanical Engineering, Kathmandu University, Dhulikhel, Nepal
| | - Iain Kilty
- Inflammation & Remodelling Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Scott Jelinsky
- Inflammation & Remodelling Research Unit, Pfizer Inc, Cambridge, MA, USA
| | - Ian P Hall
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
38
|
O'Leary C, O'Brien FJ, Cryan SA. Retinoic Acid-Loaded Collagen-Hyaluronate Scaffolds: A Bioactive Material for Respiratory Tissue Regeneration. ACS Biomater Sci Eng 2017; 3:1381-1393. [PMID: 33429696 DOI: 10.1021/acsbiomaterials.6b00561] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Clinical interventions for extensive tissue injury to the larger airways remain limited. Recently, respiratory tissue engineering strategies have emerged with a variety of biomimetic materials and tissue constructs to address these limitations, though rapid epithelialization of the construct with mucociliary function is still largely unresolved. The overall objective of this study was to manufacture an all-trans retinoic acid (atRA)-loaded bilayered collagen-hyaluronate (atRA-B) scaffold as a platform technology for tracheal tissue regeneration. atRA-loaded scaffolds were fabricated using a customized lyophilization process and characterized for drug loading and release properties using HPLC, followed by validation of their bioactivity using human primary tracheobronchial epithelial cells. atRA-loaded materials were reproducibly manufactured and exhibited the release of atRA following their hydration over 8-28 h that was significantly affected by collagen cross-linking. An optimal formulation consisting of 10 μg/mL atRA in a collagen-hyaluronate suspension to manufacture the scaffold film layer was identified and used to develop the atRA-B scaffold. Immunofluorescence studies and RT-PCR revealed that the atRA-loaded biomaterials increased the expression of two epithelial markers of mucociliary differentiation, MUC5AC and β-tubulin IV, via upregulation of MUC5AC and FOXJ1 genes, both in epithelial monoculture and in a 3D scaffold coculture system with lung fibroblasts. Overall, this study has demonstrated that the atRA-B scaffold can enhance functional epithelialization in primary tracheobronchial cells and can potentially pioneer the development of a novel and biocompatible device to address a currently unmet clinical need in tracheal replacement.
Collapse
Affiliation(s)
- Cian O'Leary
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Centre for Research in Medical Devices (CURAM), Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Centre for Research in Medical Devices (CURAM), Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.,Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Sally-Ann Cryan
- School of Pharmacy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.,Centre for Research in Medical Devices (CURAM), Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland.,Trinity Centre of Bioengineering, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| |
Collapse
|
39
|
The dangerous liaison between pollens and pollution in respiratory allergy. Ann Allergy Asthma Immunol 2017; 118:269-275. [PMID: 28143681 DOI: 10.1016/j.anai.2016.12.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/06/2016] [Accepted: 12/19/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To recapitulate the more recent epidemiologic studies on the association of air pollution with respiratory allergic diseases prevalence and to discuss the main limitations of current approaches used to establish a link between pollinosis and pollution. DATA SOURCES Through the use of PubMed, we conducted a broad literature review in the following areas: epidemiology of respiratory allergic diseases, effect of pollution and climate changes on pollen grains, and immunomodulatory properties of pollen substances. STUDY SELECTIONS Studies on short- and long-term exposure to air pollutants, such as gaseous and particulate materials, on allergic sensitization, and on exacerbation of asthma symptoms were considered. RESULTS Trend in respiratory allergic disease prevalence has increased worldwide during the last 3 decades. Although recent epidemiologic studies on a possible association of this phenomenon with increasing pollution are controversial, botanic studies suggest a clear effect of several pollutants combined to climatic changes on the increased expression of allergenic proteins in several pollen grains. The current literature suggests the need for considering both pollen allergen and pollutant contents for epidemiologic evaluation of environmental determinants in respiratory allergies. We propose that a measure of allergenic potential of pollens, indicative of the increase in allergenicity of a polluted pollen, may be considered as a new risk indicator for respiratory health in urban areas. CONCLUSION Because public greens are located in strict proximity to the anthropogenic sources of pollution, the identification of novel more reliable parameters for risk assessment in respiratory allergic diseases is an essential need for public health management and primary prevention area.
Collapse
|
40
|
Rahimi R, Htwe SS, Ochoa M, Donaldson A, Zieger M, Sood R, Tamayol A, Khademhosseini A, Ghaemmaghami AM, Ziaie B. A paper-based in vitro model for on-chip investigation of the human respiratory system. LAB ON A CHIP 2016; 16:4319-4325. [PMID: 27731881 DOI: 10.1039/c6lc00866f] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Culturing cells at the air-liquid interface (ALI) is essential for creating functional in vitro models of lung tissues. We present the use of direct-patterned laser-treated hydrophobic paper as an effective semi-permeable membrane, ideal for ALI cell culture. The surface properties of the paper are modified through a selective CO2 laser-assisted treatment to create a unique porous substrate with hydrophilic regions that regulate fluid diffusion and cell attachment. To select the appropriate model, four promising hydrophobic films were compared with each other in terms of gas permeability and long-term strength in an aqueous environment (wet-strength). Among the investigated substrates, parchment paper showed the fastest rate of oxygen permeability (3 times more than conventional transwell cell culture membranes), with the least variation in its dry and wet tensile strengths (124 MPa and 58 MPa, remaining unchanged after 7 days of submersion in PBS).The final paper-based platform provides an ideal, robust, and inexpensive device for generating monolayers of lung epithelial cells on-chip in a high-throughput fashion for disease modelling and in vitro drug testing.
Collapse
Affiliation(s)
- Rahim Rahimi
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Su Su Htwe
- Division of Immunology, School of Life Sciences, Faculty of Medicine & Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Manuel Ochoa
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| | - Amy Donaldson
- Division of Immunology, School of Life Sciences, Faculty of Medicine & Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Michael Zieger
- Indiana University School of Medicine, Division of Plastic Surgery, Indianapolis, IN, USA
| | - Rajiv Sood
- Indiana University School of Medicine, Division of Plastic Surgery, Indianapolis, IN, USA
| | - Ali Tamayol
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA and Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea and Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine & Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Babak Ziaie
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
41
|
The impact of surface chemistry modification on macrophage polarisation. Immunobiology 2016; 221:1237-46. [DOI: 10.1016/j.imbio.2016.06.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/01/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
|
42
|
Horvath P, Aulner N, Bickle M, Davies AM, Nery ED, Ebner D, Montoya MC, Östling P, Pietiäinen V, Price LS, Shorte SL, Turcatti G, von Schantz C, Carragher NO. Screening out irrelevant cell-based models of disease. Nat Rev Drug Discov 2016; 15:751-769. [PMID: 27616293 DOI: 10.1038/nrd.2016.175] [Citation(s) in RCA: 343] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The common and persistent failures to translate promising preclinical drug candidates into clinical success highlight the limited effectiveness of disease models currently used in drug discovery. An apparent reluctance to explore and adopt alternative cell- and tissue-based model systems, coupled with a detachment from clinical practice during assay validation, contributes to ineffective translational research. To help address these issues and stimulate debate, here we propose a set of principles to facilitate the definition and development of disease-relevant assays, and we discuss new opportunities for exploiting the latest advances in cell-based assay technologies in drug discovery, including induced pluripotent stem cells, three-dimensional (3D) co-culture and organ-on-a-chip systems, complemented by advances in single-cell imaging and gene editing technologies. Funding to support precompetitive, multidisciplinary collaborations to develop novel preclinical models and cell-based screening technologies could have a key role in improving their clinical relevance, and ultimately increase clinical success rates.
Collapse
Affiliation(s)
- Peter Horvath
- Synthetic and Systems Biology Unit, Biological Research Centre of the Hungarian Academy of Sciences, Szeged H-6726, Hungary; and at the Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00290, Finland.,European Cell-Based Assays Interest Group
| | - Nathalie Aulner
- Imagopole-Citech, Institut Pasteur, Paris 75015, France.,European Cell-Based Assays Interest Group
| | - Marc Bickle
- Technology Development Studio, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany.,European Cell-Based Assays Interest Group
| | - Anthony M Davies
- Translational Cell Imaging Queensland (TCIQ), Institute of Health Biomedical Innovation, Queensland University of Technology, Brisbane 4102 QLD, Australia; and The Irish National Centre for High Content Screening and Analysis, Trinity Translational Medicine Institute, Trinity College Dublin, Phase 3 Trinity Health Sciences 1.20, St James Hospital, Dublin D8, Republic of Ireland.,European Cell-Based Assays Interest Group
| | - Elaine Del Nery
- Institut Curie, PSL Research University, Department of Translational Research, The Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), F-75005, Paris, France.,European Cell-Based Assays Interest Group
| | - Daniel Ebner
- Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK.,European Cell-Based Assays Interest Group
| | - Maria C Montoya
- Cellomics Unit, Cell Biology &Physiology Program, Cell &Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.,European Cell-Based Assays Interest Group
| | - Päivi Östling
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00290, Finland.,Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institutet, Stockholm 17165, Sweden.,European Cell-Based Assays Interest Group
| | - Vilja Pietiäinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00290, Finland.,European Cell-Based Assays Interest Group
| | - Leo S Price
- Faculty of Science, Leiden Academic Centre for Drug Research, Toxicology, Universiteit Leiden, The Netherlands; and at OcellO, J.H Oortweg 21, 2333 CH, Leiden, The Netherlands.,European Cell-Based Assays Interest Group
| | - Spencer L Shorte
- Imagopole-Citech, Institut Pasteur, Paris 75015, France.,European Cell-Based Assays Interest Group
| | - Gerardo Turcatti
- Biomolecular Screening Facility, Swiss Federal Institute of Technology (EPFL), Lausanne CH-1015, Switzerland.,European Cell-Based Assays Interest Group
| | - Carina von Schantz
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00290, Finland.,European Cell-Based Assays Interest Group
| | - Neil O Carragher
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XR, UK.,European Cell-Based Assays Interest Group
| |
Collapse
|
43
|
Wiles K, Fishman JM, De Coppi P, Birchall MA. The Host Immune Response to Tissue-Engineered Organs: Current Problems and Future Directions. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:208-19. [DOI: 10.1089/ten.teb.2015.0376] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | | | | | - Martin A. Birchall
- UCL Ear Institute & Royal National Throat, Nose and Ear Hospital, London, United Kingdom
| |
Collapse
|
44
|
Marrazzo P, Maccari S, Taddei A, Bevan L, Telford J, Soriani M, Pezzicoli A. 3D Reconstruction of the Human Airway Mucosa In Vitro as an Experimental Model to Study NTHi Infections. PLoS One 2016; 11:e0153985. [PMID: 27101006 PMCID: PMC4839639 DOI: 10.1371/journal.pone.0153985] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/06/2016] [Indexed: 12/04/2022] Open
Abstract
We have established an in vitro 3D system which recapitulates the human tracheo-bronchial mucosa comprehensive of the pseudostratified epithelium and the underlying stromal tissue. In particular, we reported that the mature model, entirely constituted of primary cells of human origin, develops key markers proper of the native tissue such as the mucociliary differentiation of the epithelial sheet and the formation of the basement membrane. The infection of the pseudo-tissue with a strain of NonTypeable Haemophilus influenzae results in bacteria association and crossing of the mucus layer leading to an apparent targeting of the stromal space where they release large amounts of vesicles and form macro-structures. In summary, we propose our in vitro model as a reliable and potentially customizable system to study mid/long term host-pathogen processes.
Collapse
Affiliation(s)
| | - Silvia Maccari
- GSK Vaccines S.r.l., via Fiorentina 1, 53100, Siena, Italy
| | - Annarita Taddei
- Interdepartmental Centre for Electron Microscopy, Tuscia University, Viterbo, Italy
| | - Luke Bevan
- Respiratory Disease Area, Novartis Institutes for BioMedical Research, Horsham, RH12 5AB, United Kingdom
| | - John Telford
- GSK Vaccines S.r.l., via Fiorentina 1, 53100, Siena, Italy
| | - Marco Soriani
- GSK Vaccines S.r.l., via Fiorentina 1, 53100, Siena, Italy
- * E-mail: (AP); (MS)
| | - Alfredo Pezzicoli
- GSK Vaccines S.r.l., via Fiorentina 1, 53100, Siena, Italy
- * E-mail: (AP); (MS)
| |
Collapse
|
45
|
The development of a tissue-engineered tracheobronchial epithelial model using a bilayered collagen-hyaluronate scaffold. Biomaterials 2016; 85:111-27. [PMID: 26871888 DOI: 10.1016/j.biomaterials.2016.01.065] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 01/25/2016] [Accepted: 01/28/2016] [Indexed: 02/05/2023]
Abstract
Today, chronic respiratory disease is one of the leading causes of mortality globally. Epithelial dysfunction can play a central role in its pathophysiology. The development of physiologically-representative in vitro model systems using tissue-engineered constructs might improve our understanding of epithelial tissue and disease. This study sought to engineer a bilayered collagen-hyaluronate (CHyA-B) scaffold for the development of a physiologically-representative 3D in vitro tracheobronchial epithelial co-culture model. CHyA-B scaffolds were fabricated by integrating a thin film top-layer into a porous sub-layer with lyophilisation. The film layer firmly connected to the sub-layer with delamination occurring at stresses of 12-15 kPa. Crosslinked scaffolds had a compressive modulus of 1.9 kPa and mean pore diameters of 70 μm and 80 μm, depending on the freezing temperature. Histological analysis showed that the Calu-3 bronchial epithelial cell line attached and grew on CHyA-B with adoption of an epithelial monolayer on the film layer. Immunofluorescence and qRT-PCR studies demonstrated that the CHyA-B scaffolds facilitated Calu-3 cell differentiation, with enhanced mucin expression, increased ciliation and the formation of intercellular tight junctions. Co-culture of Calu-3 cells with Wi38 lung fibroblasts was achieved on the scaffold to create a submucosal tissue analogue of the upper respiratory tract, validating CHyA-B as a platform to support co-culture and cellular organisation reminiscent of in vivo tissue architecture. In summary, this study has demonstrated that CHyA-B is a promising tool for the development of novel 3D tracheobronchial co-culture in vitro models with the potential to unravel new pathways in drug discovery and drug delivery.
Collapse
|
46
|
Ghadiri M, Young PM, Traini D. Cell-based therapies for the treatment of idiopathic pulmonary fibrosis (IPF) disease. Expert Opin Biol Ther 2015; 16:375-87. [PMID: 26593230 DOI: 10.1517/14712598.2016.1124085] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION During the last few decades, cell-based therapies have shown great potential to treat patients with lung diseases. It has been proposed that the administration of cells into an injured lung could be considered as a therapeutic method to repair and replace lost lung tissue. Using this method, transplanted cells with the ability to proliferate and differentiate into alveolar cells, have been suggested as a therapeutic strategy for IPF treatment. AREAS COVERED In this review, the latest investigations using various types of cells for IPF therapy have been presented. The cells studied for cell-based therapies in IPF are lung alveolar epithelial cells, lung resident stem cells and exogenous adult stem cells such as MSCs. EXPERT OPINION After many years of investigation, the use of cell-based therapies to treat IPF is still at the experimental phase. Problems include bioethical issues, safety of cell transplantation, routes of delivery and the dose and timing of administration. Further investigations are necessary to establish the best strategy for using cell-based therapies effectively for the treatment of IPF.
Collapse
Affiliation(s)
- Maliheh Ghadiri
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Paul M Young
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| | - Daniela Traini
- a Respiratory Technology, Woolcock Institute of Medical Research and Discipline of Pharmacology , Sydney Medical School , Sydney , NSW , Australia
| |
Collapse
|
47
|
Htwe SS, Harrington H, Knox A, Rose F, Aylott J, Haycock JW, Ghaemmaghami AM. Investigating NF-κB signaling in lung fibroblasts in 2D and 3D culture systems. Respir Res 2015; 16:144. [PMID: 26619903 PMCID: PMC4666055 DOI: 10.1186/s12931-015-0302-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/13/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Inflammatory respiratory diseases are amongst major global health challenges. Lung fibroblasts have been shown to play a key role in lung inflammatory responses. However, their exact role in initiation and maintenance of lung diseases has remained elusive partly due to the limited availability of physiologically relevant in vitro models. Therefore, developing new tools that enable investigating the molecular pathways (e.g. nuclear factor-kappa B (NF-κB) activation) that underpin inflammatory responses in fibroblasts could be a valuable resource for scientists working in this area of research. RESULTS In order to investigate NF-κB activation in response to pro-inflammatory stimuli in real-time, we first developed two detection systems based on nuclear localization of NF-κB by immunostaining and luciferase reporter assay system. Furthermore using electrospun porous scaffolds, with similar geometry to human lung extracellular matrix, we developed 3D cultures of lung fibroblasts allowing comparing NF-κB activation in response to pro-inflammatory stimuli (i.e. TNF-α) in 2D and 3D. Our data clearly show that the magnitude of NF-κB activation in 2D cultures is substantially higher than 3D cultures. However, unlike 2D cultures, cells in the 3D model remained responsive to TNF-α at higher concentrations. The more subdued and wider dynamic range of NF-κB responses in 3D culture system was associated with a different expression pattern for TNF receptor I in 3D versus 2D cultures collectively reflecting a more in vivo like TNF receptor I expression and NF-κB activation pattern in the 3D system. CONCLUSION Our data suggest that lung fibroblasts are actively involved in the pathogenesis of lung inflammation by activation of NF-κB signaling pathway. The 3D culture detection system provides a sensitive and biologically relevant tool for investigating different pro-inflammatory events involving lung fibroblasts.
Collapse
Affiliation(s)
- Su Su Htwe
- Cellular Immunology and Allergy Research Group, Division of Immunology, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK.
| | - Helen Harrington
- Cellular Immunology and Allergy Research Group, Division of Immunology, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK.
| | - Alan Knox
- Division of Respiratory Medicine, University of Nottingham, City Hospital, Nottingham, UK.
| | - Felicity Rose
- Division of Drug Delivery and Tissue Engineering, Centre for Biomolecular Sciences, School of Pharmacy, University of Nottingham, Nottingham, UK.
| | - Jonathan Aylott
- Laboratory of Biophysics and Surface Analysis, School of Pharmacy, University of Nottingham, Nottingham, UK.
| | - John W Haycock
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK.
| | - Amir M Ghaemmaghami
- Cellular Immunology and Allergy Research Group, Division of Immunology, School of Life Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
48
|
Zhang H, Fu W, Xu Z. Re-epithelialization: a key element in tracheal tissue engineering. Regen Med 2015; 10:1005-23. [PMID: 26388452 DOI: 10.2217/rme.15.68] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Trachea-tissue engineering is a thriving new field in regenerative medicine that is reaching maturity and yielding numerous promising results. In view of the crucial role that the epithelium plays in the trachea, re-epithelialization of tracheal substitutes has gradually emerged as the focus of studies in tissue-engineered trachea. Recent progress in our understanding of stem cell biology, growth factor interactions and transplantation immunobiology offer the prospect of optimization of a tissue-engineered tracheal epithelium. In addition, advances in cell culture technology and successful applications of clinical transplantation are opening up new avenues for the construction of a tissue-engineered tracheal epithelium. Therefore, this review summarizes current advances, unresolved obstacles and future directions in the reconstruction of a tissue-engineered tracheal epithelium.
Collapse
Affiliation(s)
- Hengyi Zhang
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China.,Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| | - Zhiwei Xu
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, China
| |
Collapse
|
49
|
Fujie T, Shi X, Ostrovidov S, Liang X, Nakajima K, Chen Y, Wu H, Khademhosseini A. Spatial coordination of cell orientation directed by nanoribbon sheets. Biomaterials 2015; 53:86-94. [PMID: 25890709 DOI: 10.1016/j.biomaterials.2015.02.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/02/2015] [Indexed: 01/05/2023]
Abstract
Spatial coordination of cell orientation is of central importance in tissue/organ construction. In this study, we developed microfabricated poly(lactic-co-glycolic acid) (PLGA) nanoribbon sheets with unique structures, using spin-coating and micropatterning techniques, in order to generate a hierarchically assembled cellular structure consisting of murine skeletal myoblasts (C2C12). The nanoribbon sheets were composed of aligned PLGA nanoribbons in the center, and strips on four sides which take a role as bridges to connect and immobilize the aligned nanoribbons. Such unique structures facilitated the alignment of C2C12 cells into bilayer cell sheets, and cellular alignment was directed by the aligned direction of nanoribbons. The nanoribbon sheets also facilitated the construction of multilayer cell sheets with anisotropic (orthogonal) and isotropic (parallel) orientations. The enhanced expression of myogenic genes of C2C12 cells on the bilayer cell sheets demonstrated that the nanoribbons induced C2C12 cell differentiation into mature myoblasts. The micropatterned nanoribbon sheets may be a useful tool for directing cellular organization with defined alignment for regenerative medicine and drug screening applications.
Collapse
Affiliation(s)
- Toshinori Fujie
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Xuetao Shi
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan
| | - Serge Ostrovidov
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan
| | - Xiaobin Liang
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan
| | - Ken Nakajima
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan
| | - Yin Chen
- Department of Chemistry & Division of Biomedical Engineering, Hong Kong University of Science & Technology, Hong Kong, China
| | - Hongkai Wu
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan; Department of Chemistry & Division of Biomedical Engineering, Hong Kong University of Science & Technology, Hong Kong, China.
| | - Ali Khademhosseini
- WPI-Advanced Institute for Materials Research, Tohoku University, Sendai 980-8578, Japan; Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|
50
|
O'Leary C, Gilbert JL, O'Dea S, O'Brien FJ, Cryan SA. Respiratory Tissue Engineering: Current Status and Opportunities for the Future. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:323-44. [PMID: 25587703 DOI: 10.1089/ten.teb.2014.0525] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Currently, lung disease and major airway trauma constitute a major global healthcare burden with limited treatment options. Airway diseases such as chronic obstructive pulmonary disease and cystic fibrosis have been identified as the fifth highest cause of mortality worldwide and are estimated to rise to fourth place by 2030. Alternate approaches and therapeutic modalities are urgently needed to improve clinical outcomes for chronic lung disease. This can be achieved through tissue engineering of the respiratory tract. Interest is growing in the use of airway tissue-engineered constructs as both a research tool, to further our understanding of airway pathology, validate new drugs, and pave the way for novel drug therapies, and also as regenerative medical devices or as an alternative to transplant tissue. This review provides a concise summary of the field of respiratory tissue engineering to date. An initial overview of airway anatomy and physiology is given, followed by a description of the stem cell populations and signaling processes involved in parenchymal healing and tissue repair. We then focus on the different biomaterials and tissue-engineered systems employed in upper and lower respiratory tract engineering and give a final perspective of the opportunities and challenges facing the field of respiratory tissue engineering.
Collapse
Affiliation(s)
- Cian O'Leary
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,2 School of Pharmacy, Royal College of Surgeons in Ireland , Dublin, Ireland .,3 Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin , Dublin, Ireland
| | - Jennifer L Gilbert
- 4 Department of Biology, Institute of Immunology, University of Ireland , Maynooth, Ireland
| | - Shirley O'Dea
- 4 Department of Biology, Institute of Immunology, University of Ireland , Maynooth, Ireland
| | - Fergal J O'Brien
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,3 Advanced Materials and Bioengineering Research (AMBER) Centre, Royal College of Surgeons in Ireland and Trinity College Dublin , Dublin, Ireland .,5 Trinity Centre of Bioengineering, Trinity College Dublin , Dublin, Ireland
| | - Sally-Ann Cryan
- 1 Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland , Dublin, Ireland .,2 School of Pharmacy, Royal College of Surgeons in Ireland , Dublin, Ireland .,5 Trinity Centre of Bioengineering, Trinity College Dublin , Dublin, Ireland
| |
Collapse
|