51
|
Xiao H, Nguyen RY, LaRanger R, Herzog EL, Mak M. Integrated computational and experimental pipeline for quantifying local cell-matrix interactions. Sci Rep 2021; 11:16465. [PMID: 34385554 PMCID: PMC8361134 DOI: 10.1038/s41598-021-95935-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 07/31/2021] [Indexed: 11/30/2022] Open
Abstract
Cellular interactions with the extracellular matrix (ECM) play a key role in modulating biological processes. While studies have identified key molecular factors of these interactions, the mechanical regulation associated with these interactions is not well characterized. To address this, we present an image analysis platform to analyze time-dependent dynamics observed in lung fibroblasts embedded in a 3D collagen matrix. Combining drug studies with quantitative analysis of cell–matrix interactions, our results are able to provide cellular level quantitative insights for mechanical and biophysical phenomena relevant to cell-ECM interactions. This system overall represents an initial pipeline for understanding cell mechanics in a 3D collagen gel and their implications in a physiologically relevant context.
Collapse
Affiliation(s)
- Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Ryan LaRanger
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Erica L Herzog
- Department of Medicine (Pulmonary, Critical Care and Sleep), Yale University School of Medicine, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
52
|
Lagares D, Hinz B. Animal and Human Models of Tissue Repair and Fibrosis: An Introduction. Methods Mol Biol 2021; 2299:277-290. [PMID: 34028750 DOI: 10.1007/978-1-0716-1382-5_20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reductionist cell culture systems are not only convenient but essential to understand molecular mechanisms of myofibroblast activation and action in carefully controlled conditions. However, tissue myofibroblasts do not act in isolation and the complexity of tissue repair and fibrosis in humans cannot be captured even by the most elaborate culture models. Over the past five decades, numerous animal models have been developed to study different aspects of myofibroblast biology and interactions with other cells and extracellular matrix. The underlying principles can be broadly classified into: (1) organ injury by trauma such as prototypical full thickness skin wounds or burns; (2) mechanical challenges, such as pressure overload of the heart by ligature of the aorta or the pulmonary vein; (3) toxic injury, such as administration of bleomycin to lungs and carbon tetrachloride to the liver; (4) organ infection with viruses, bacteria, and parasites, such as nematode infections of liver; (5) cytokine and inflammatory models, including local delivery or viral overexpression of active transforming growth factor beta; (6) "lifestyle" and metabolic models such as high-fat diet; and (7) various genetic models. We will briefly summarize the most widely used mouse models used to study myofibroblasts in tissue repair and fibrosis as well as genetic tools for manipulating myofibroblast repair functions in vivo.
Collapse
Affiliation(s)
- David Lagares
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
53
|
Xu Y, Koya R, Ask K, Zhao R. Engineered microenvironment for the study of myofibroblast mechanobiology. Wound Repair Regen 2021; 29:588-596. [PMID: 34118169 PMCID: PMC8254796 DOI: 10.1111/wrr.12955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
Myofibroblasts are mechanosensitive cells and a variety of their behaviours including differentiation, migration, force production and biosynthesis are regulated by the surrounding microenvironment. Engineered cell culture models have been developed to examine the effect of microenvironmental factors such as the substrate stiffness, the topography and strain of the extracellular matrix (ECM) and the shear stress on myofibroblast biology. These engineered models provide well-mimicked, pathophysiologically relevant experimental conditions that are superior to those enabled by the conventional two-dimensional (2D) culture models. In this perspective, we will review the recent advances in the development of engineered cell culture models for myofibroblasts and outline the findings on the myofibroblast mechanobiology under various microenvironmental conditions. These studies have demonstrated the power and utility of the engineered models for the study of microenvironment-regulated cellular behaviours. The findings derived using these models contribute to a greater understanding of how myofibroblast behaviour is regulated in tissue repair and pathological scar formation.
Collapse
Affiliation(s)
- Ying Xu
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - Richard Koya
- Department of Obstetrics and Gynecology, University of Chicago Comprehensive Cancer Center, Biological Sciences Division, University of Chicago School of Medicine, Chicago, IL 60637, USA
| | - Kjetil Ask
- Department of Medicine, Div. Respirology, McMaster University, Hamilton, ON, Canada L8N 4A6
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, Hamilton, ON, Canada L8N 4A6
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
54
|
Sun AM, Hoffman T, Luu BQ, Ashammakhi N, Li S. Application of lung microphysiological systems to COVID-19 modeling and drug discovery: a review. Biodes Manuf 2021; 4:757-775. [PMID: 34178414 PMCID: PMC8213042 DOI: 10.1007/s42242-021-00136-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
There is a pressing need for effective therapeutics for coronavirus disease 2019 (COVID-19), the respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. The process of drug development is a costly and meticulously paced process, where progress is often hindered by the failure of initially promising leads. To aid this challenge, in vitro human microphysiological systems need to be refined and adapted for mechanistic studies and drug screening, thereby saving valuable time and resources during a pandemic crisis. The SARS-CoV-2 virus attacks the lung, an organ where the unique three-dimensional (3D) structure of its functional units is critical for proper respiratory function. The in vitro lung models essentially recapitulate the distinct tissue structure and the dynamic mechanical and biological interactions between different cell types. Current model systems include Transwell, organoid and organ-on-a-chip or microphysiological systems (MPSs). We review models that have direct relevance toward modeling the pathology of COVID-19, including the processes of inflammation, edema, coagulation, as well as lung immune function. We also consider the practical issues that may influence the design and fabrication of MPS. The role of lung MPS is addressed in the context of multi-organ models, and it is discussed how high-throughput screening and artificial intelligence can be integrated with lung MPS to accelerate drug development for COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Argus M. Sun
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- UC San Diego Healthcare, UCSD, La Jolla, CA 92037 USA
| | - Tyler Hoffman
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
| | - Bao Q. Luu
- Pulmonary Diseases and Critical Care, Scripps Green Hospital, Scripps Health, La Jolla, CA 92037 USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824 USA
| | - Song Li
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 USA
| |
Collapse
|
55
|
Ptasinski V, Stegmayr J, Belvisi MG, Wagner DE, Murray LA. Targeting Alveolar Repair in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 65:347-365. [PMID: 34129811 PMCID: PMC8525210 DOI: 10.1165/rcmb.2020-0476tr] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal interstitial lung disease with limited therapeutic options. Current evidence suggests that IPF may be initiated by repeated epithelial injury in the distal lung followed by abnormal wound healing responses which occur due to intrinsic and extrinsic factors. Mechanisms contributing to chronic damage of the alveolar epithelium in IPF include dysregulated cellular processes such as apoptosis, senescence, abnormal activation of developmental pathways, aging, as well as genetic mutations. Therefore, targeting the regenerative capacity of the lung epithelium is an attractive approach in the development of novel therapies for IPF. Endogenous lung regeneration is a complex process involving coordinated cross-talk between multiple cell types and re-establishment of a normal extracellular matrix environment. This review will describe the current knowledge of reparative epithelial progenitor cells in the alveolar region of the lung and discuss potential novel therapeutic approaches for IPF focusing on endogenous alveolar repair. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Collapse
Affiliation(s)
- Victoria Ptasinski
- Lund University Faculty of Medicine, 59568, Lund, Sweden.,AstraZeneca R&D Gothenburg, 128698, Goteborg, Sweden
| | - John Stegmayr
- Lunds University Faculty of Medicine, 59568, Lund, Sweden
| | - Maria G Belvisi
- Imperial College London, 4615, London, United Kingdom of Great Britain and Northern Ireland
| | - Darcy E Wagner
- Lunds Universitet, 5193, Experimental Medical Sciences, Lund, Sweden
| | - Lynne A Murray
- AstraZeneca PLC, 4625, Cambridge, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
56
|
Kuno H, Akagi T, Fukui E, Yamato H, Akashi M, Shintani Y. Three-Dimensional Idiopathic Pulmonary Fibrosis Model Using a Layer-by-Layer Cell Coating Technique. Tissue Eng Part C Methods 2021; 27:378-390. [PMID: 34074128 DOI: 10.1089/ten.tec.2020.0365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a severe health problem characterized by progressive fibroblast proliferation and aberrant vascular remodeling. However, the lack of a suitable in vitro model that replicates cell-specific changes in IPF tissue is a crucial issue. Three-dimensional (3D) cell cultures allow the mimicking of cell-specific functions, facilitating development of novel antifibrosis drugs. We have established a layer-by-layer (LbL) cell coating technique that enables the construction of 3D tissue and also vascularized 3D tissue. This study evaluated whether this technique is beneficial for constructing an in vitro IPF-3D model using human lung fibroblasts and microvascular endothelial cells. We fabricated an in vitro IPF-3D model to provide IPF-derived fibroblasts-specific function and aberrant microvascular structure using the LbL cell coating technique. We also found that this in vitro IPF-3D model showed drug responsiveness to two antifibrosis drugs that have recently been approved worldwide. This in vitro IPF-3D model constructed by a LbL cell coating technique would help in the understanding of fibroblast function and the microvascular environment in IPF and could also be used to predict the efficacy of novel antifibrosis drugs. Impact statement We established a novel in vitro model mimicking idiopathic pulmonary fibrosis. Three-dimensional culture was constructed by layer-by-layer cell coating technique. This novel model provides a visualization of fibroblast-specific function. This assay allows for the assessment of pulmonary microvascular environment. Our model may be useful for predicting the efficacy of novel antifibrosis drugs.
Collapse
Affiliation(s)
- Hidenori Kuno
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takami Akagi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Eriko Fukui
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiroyuki Yamato
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Mitsuru Akashi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
57
|
Ort C, Chen Y, Ghagre A, Ehrlicher A, Moraes C. Bioprintable, Stiffness-Tunable Collagen-Alginate Microgels for Increased Throughput 3D Cell Culture Studies. ACS Biomater Sci Eng 2021; 7:2814-2822. [PMID: 34019377 DOI: 10.1021/acsbiomaterials.1c00129] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
3D culture platforms with tunable stiffness have the potential to improve many applications, such as drug discovery, organoid studies, and stem cell differentiation. Both dimensionality and stiffness regulate crucial and relevant cellular processes. However, 3D culture models are often limited in throughput and difficult to adopt for widespread use. Here, we demonstrate an accessible 3D, stiffness-tunable tissue culture platform, based on an interpenetrating network of collagen-1 and alginate. When blended with polymers that induce phase separation, these networks can be bioprinted at microliter volumes, using standard liquid handling infrastructure. We demonstrate robust reproducibility in printing these microgels, consistent tunability of mechanical properties, and maintained viability of multiple printed cell types. To highlight the utility and importance of this system, we demonstrate distinct morphological changes to cells in culture, use the system to probe the role of matrix mechanics and soluble factors in a collagen contraction assay, and perform a prototype viability screen against a candidate chemotherapeutic, demonstrating stiffness-dependent responses.
Collapse
Affiliation(s)
- Carley Ort
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada
| | - Yimai Chen
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal H3A 2K6, Quebec, Canada
| | - Allen Ehrlicher
- Department of Biomedical Engineering, McGill University, 3775 rue University, Montreal H3A 2B4, Quebec, Canada.,Department of Bioengineering, McGill University, 817 Sherbrooke Street West, Montreal H3A 2K6, Quebec, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal H3A 1A3, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Quebec, Canada.,Department of Mechanical Engineering, McGill University, Montreal H3A 0C3, Quebec, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, 3610 rue University, Montreal H3A 0G4, Quebec, Canada.,Department of Biomedical Engineering, McGill University, 3775 rue University, Montreal H3A 2B4, Quebec, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal H3A 1A3, Quebec, Canada
| |
Collapse
|
58
|
Kiener M, Roldan N, Machahua C, Sengupta A, Geiser T, Guenat OT, Funke-Chambour M, Hobi N, Kruithof-de Julio M. Human-Based Advanced in vitro Approaches to Investigate Lung Fibrosis and Pulmonary Effects of COVID-19. Front Med (Lausanne) 2021; 8:644678. [PMID: 34026781 PMCID: PMC8139419 DOI: 10.3389/fmed.2021.644678] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused considerable socio-economic burden, which fueled the development of treatment strategies and vaccines at an unprecedented speed. However, our knowledge on disease recovery is sparse and concerns about long-term pulmonary impairments are increasing. Causing a broad spectrum of symptoms, COVID-19 can manifest as acute respiratory distress syndrome (ARDS) in the most severely affected patients. Notably, pulmonary infection with Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2), the causing agent of COVID-19, induces diffuse alveolar damage (DAD) followed by fibrotic remodeling and persistent reduced oxygenation in some patients. It is currently not known whether tissue scaring fully resolves or progresses to interstitial pulmonary fibrosis. The most aggressive form of pulmonary fibrosis is idiopathic pulmonary fibrosis (IPF). IPF is a fatal disease that progressively destroys alveolar architecture by uncontrolled fibroblast proliferation and the deposition of collagen and extracellular matrix (ECM) proteins. It is assumed that micro-injuries to the alveolar epithelium may be induced by inhalation of micro-particles, pathophysiological mechanical stress or viral infections, which can result in abnormal wound healing response. However, the exact underlying causes and molecular mechanisms of lung fibrosis are poorly understood due to the limited availability of clinically relevant models. Recently, the emergence of SARS-CoV-2 with the urgent need to investigate its pathogenesis and address drug options, has led to the broad application of in vivo and in vitro models to study lung diseases. In particular, advanced in vitro models including precision-cut lung slices (PCLS), lung organoids, 3D in vitro tissues and lung-on-chip (LOC) models have been successfully employed for drug screens. In order to gain a deeper understanding of SARS-CoV-2 infection and ultimately alveolar tissue regeneration, it will be crucial to optimize the available models for SARS-CoV-2 infection in multicellular systems that recapitulate tissue regeneration and fibrotic remodeling. Current evidence for SARS-CoV-2 mediated pulmonary fibrosis and a selection of classical and novel lung models will be discussed in this review.
Collapse
Affiliation(s)
- Mirjam Kiener
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Nuria Roldan
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Carlos Machahua
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arunima Sengupta
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Olivier Thierry Guenat
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research DBMR, Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nina Hobi
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Department for BioMedical Research DBMR, Urology Research Laboratory, University of Bern, Bern, Switzerland
- Alveolix AG, Swiss Organs-on-Chip Innovation, Bern, Switzerland
- Organoid Core, Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
59
|
Anandakrishnan N, Ye H, Guo Z, Chen Z, Mentkowski KI, Lang JK, Rajabian N, Andreadis ST, Ma Z, Spernyak JA, Lovell JF, Wang D, Xia J, Zhou C, Zhao R. Fast Stereolithography Printing of Large-Scale Biocompatible Hydrogel Models. Adv Healthc Mater 2021; 10:e2002103. [PMID: 33586366 PMCID: PMC8212355 DOI: 10.1002/adhm.202002103] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 11/07/2022]
Abstract
Large size cell-laden hydrogel models hold great promise for tissue repair and organ transplantation, but their fabrication using 3D bioprinting is limited by the slow printing speed that can affect the part quality and the biological activity of the encapsulated cells. Here a fast hydrogel stereolithography printing (FLOAT) method is presented that allows the creation of a centimeter-sized, multiscale solid hydrogel model within minutes. Through precisely controlling the photopolymerization condition, low suction force-driven, high-velocity flow of the hydrogel prepolymer is established that supports the continuous replenishment of the prepolymer solution below the curing part and the nonstop part growth. The rapid printing of centimeter-sized hydrogel models using FLOAT is shown to significantly reduce the part deformation and cellular injury caused by the prolonged exposure to the environmental stresses in conventional 3D printing methods. Embedded vessel networks fabricated through multiscale printing allows media perfusion needed to maintain the high cellular viability and metabolic functions in the deep core of the large-sized models. The endothelialization of this vessel network allows the establishment of barrier functions. Together, these studies demonstrate a rapid 3D hydrogel printing method and represent a first step toward the fabrication of large-sized engineered tissue models.
Collapse
Affiliation(s)
- Nanditha Anandakrishnan
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Hang Ye
- Department of Industrial and Systems Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Zipeng Guo
- Department of Industrial and Systems Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Zhaowei Chen
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Kyle I Mentkowski
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Jennifer K Lang
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
- VA WNY Healthcare System, Buffalo, NY, 14215, USA
| | - Nika Rajabian
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Stelios T Andreadis
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Zhen Ma
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Joseph A Spernyak
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Depeng Wang
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Jun Xia
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Chi Zhou
- Department of Industrial and Systems Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ruogang Zhao
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| |
Collapse
|
60
|
De Pieri A, Korman BD, Jüngel A, Wuertz-Kozak K. Engineering Advanced In Vitro Models of Systemic Sclerosis for Drug Discovery and Development. Adv Biol (Weinh) 2021; 5:e2000168. [PMID: 33852183 PMCID: PMC8717409 DOI: 10.1002/adbi.202000168] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a complex multisystem disease with the highest case-specific mortality among all autoimmune rheumatic diseases, yet without any available curative therapy. Therefore, the development of novel therapeutic antifibrotic strategies that effectively decrease skin and organ fibrosis is needed. Existing animal models are cost-intensive, laborious and do not recapitulate the full spectrum of the disease and thus commonly fail to predict human efficacy. Advanced in vitro models, which closely mimic critical aspects of the pathology, have emerged as valuable platforms to investigate novel pharmaceutical therapies for the treatment of SSc. This review focuses on recent advancements in the development of SSc in vitro models, sheds light onto biological (e.g., growth factors, cytokines, coculture systems), biochemical (e.g., hypoxia, reactive oxygen species) and biophysical (e.g., stiffness, topography, dimensionality) cues that have been utilized for the in vitro recapitulation of the SSc microenvironment, and highlights future perspectives for effective drug discovery and validation.
Collapse
Affiliation(s)
- Andrea De Pieri
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
| | - Benjamin D Korman
- Prof. B. D. Korman, Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Astrid Jüngel
- Prof. A. Jüngel, Center of Experimental Rheumatology, University Clinic of Rheumatology, Balgrist University Hospital, University Hospital Zurich, Zurich, 8008, Switzerland
- Prof. A. Jüngel, Department of Physical Medicine and Rheumatology, Balgrist University Hospital, University of Zurich, Zurich, 8008, Switzerland
| | - Karin Wuertz-Kozak
- Dr. A. De Pieri, Prof. K. Wuertz-Kozak, Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 106 Lomb Memorial Rd., Rochester, NY, 14623, USA
- Prof. K. Wuertz-Kozak, Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), Munich, 81547, Germany
| |
Collapse
|
61
|
Nanofiber Configuration of Electrospun Scaffolds Dictating Cell Behaviors and Cell-scaffold Interactions. Chem Res Chin Univ 2021. [DOI: 10.1007/s40242-021-1024-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
62
|
Gracioso Martins AM, Wilkins MD, Ligler FS, Daniele MA, Freytes DO. Microphysiological System for High-Throughput Computer Vision Measurement of Microtissue Contraction. ACS Sens 2021; 6:985-994. [PMID: 33656335 DOI: 10.1021/acssensors.0c02172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The ability to measure microtissue contraction in vitro can provide important information when modeling cardiac, cardiovascular, respiratory, digestive, dermal, and skeletal tissues. However, measuring tissue contraction in vitro often requires the use of high number of cells per tissue construct along with time-consuming microscopy and image analysis. Here, we present an inexpensive, versatile, high-throughput platform to measure microtissue contraction in a 96-well plate configuration using one-step batch imaging. More specifically, optical fiber microprobes are embedded in microtissues, and contraction is measured as a function of the deflection of optical signals emitted from the end of the fibers. Signals can be measured from all the filled wells on the plate simultaneously using a digital camera. An algorithm uses pixel-based image analysis and computer vision techniques for the accurate multiwell quantification of positional changes in the optical microprobes caused by the contraction of the microtissues. Microtissue constructs containing 20,000-100,000 human ventricular cardiac fibroblasts (NHCF-V) in 6 mg/mL collagen type I showed contractile displacements ranging from 20-200 μm. This highly sensitive and versatile platform can be used for the high-throughput screening of microtissues in disease modeling, drug screening for therapeutics, physiology research, and safety pharmacology.
Collapse
Affiliation(s)
- Ana Maria Gracioso Martins
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill/North Carolina State University, Raleigh 27695, North Carolina, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh 27695, North Carolina, United States
| | - Michael D. Wilkins
- Comparative Medicine Institute, North Carolina State University, Raleigh 27695, North Carolina, United States
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh 27695, North Carolina, United States
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill/North Carolina State University, Raleigh 27695, North Carolina, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh 27695, North Carolina, United States
| | - Michael A. Daniele
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill/North Carolina State University, Raleigh 27695, North Carolina, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh 27695, North Carolina, United States
- Department of Electrical and Computer Engineering, North Carolina State University, Raleigh 27695, North Carolina, United States
| | - Donald O. Freytes
- Joint Department of Biomedical Engineering, University of North Carolina-Chapel Hill/North Carolina State University, Raleigh 27695, North Carolina, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh 27695, North Carolina, United States
| |
Collapse
|
63
|
Zamprogno P, Wüthrich S, Achenbach S, Thoma G, Stucki JD, Hobi N, Schneider-Daum N, Lehr CM, Huwer H, Geiser T, Schmid RA, Guenat OT. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol 2021; 4:168. [PMID: 33547387 PMCID: PMC7864995 DOI: 10.1038/s42003-021-01695-0] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Simon Wüthrich
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Sven Achenbach
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Janick D Stucki
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nina Hobi
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nicole Schneider-Daum
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Hanno Huwer
- SHG Clinics, Department of Cardiothoracic Surgery, Völklingen Heart Center, Völklingen, Germany
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland.
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
64
|
Calcagno TM, Zhang C, Tian R, Ebrahimi B, Mirsaeidi M. Novel three-dimensional biochip pulmonary sarcoidosis model. PLoS One 2021; 16:e0245805. [PMID: 33539409 PMCID: PMC7861546 DOI: 10.1371/journal.pone.0245805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcoidosis is a multi-system disorder of granulomatous inflammation which most commonly affects the lungs. Its etiology and pathogenesis are not well defined in part due to the lack of reliable modeling. Here, we present the development of an in vitro three-dimensional lung-on-chip biochip designed to mimic granuloma formation. A lung on chip fluidic macrodevice was developed and added to our previously developed a lung-on-membrane model (LOMM). Granulomas were cultured from blood samples of patients with sarcoidosis and then inserted in the air-lung-interface of the microchip to create a three-dimensional biochip pulmonary sarcoidosis model (3D BSGM). Cytokines were measured after 48 hours. ELISA testing was performed to measure cytokine response difference between LOMM with 3D BSGM. There were statistically significant differences in IL-1ß (P = 0.001953), IL-6 (P = 0.001953), GM-CSF (P = 0.001953), and INF-γ expressions (P = 0.09375) between two groups. The current model represents the first 3D biochip sarcoidosis model created by adding a microfluidics system to a dual-chambered lung on membrane model and introducing developed sarcoid-granuloma to its air-lung-interface.
Collapse
Affiliation(s)
- Tess M. Calcagno
- Department of Medicine, University of Miami, Miami, FL, United States of America
| | - Chongxu Zhang
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States of America
| | - Runxia Tian
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States of America
| | - Babak Ebrahimi
- Research and Development, Genix-Engineering, Irvin, California, United States of America
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
65
|
Kang SM, Kim D, Lee JH, Takayama S, Park JY. Engineered Microsystems for Spheroid and Organoid Studies. Adv Healthc Mater 2021; 10:e2001284. [PMID: 33185040 PMCID: PMC7855453 DOI: 10.1002/adhm.202001284] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/01/2020] [Indexed: 01/09/2023]
Abstract
3D in vitro model systems such as spheroids and organoids provide an opportunity to extend the physiological understanding using recapitulated tissues that mimic physiological characteristics of in vivo microenvironments. Unlike 2D systems, 3D in vitro systems can bridge the gap between inadequate 2D cultures and the in vivo environments, providing novel insights on complex physiological mechanisms at various scales of organization, ranging from the cellular, tissue-, to organ-levels. To satisfy the ever-increasing need for highly complex and sophisticated systems, many 3D in vitro models with advanced microengineering techniques have been developed to answer diverse physiological questions. This review summarizes recent advances in engineered microsystems for the development of 3D in vitro model systems. The relationship between the underlying physics behind the microengineering techniques, and their ability to recapitulate distinct 3D cellular structures and functions of diverse types of tissues and organs are highlighted and discussed in detail. A number of 3D in vitro models and their engineering principles are also introduced. Finally, current limitations are summarized, and perspectives for future directions in guiding the development of 3D in vitro model systems using microengineering techniques are provided.
Collapse
Affiliation(s)
- Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan, Chungnam, 31066, Republic of Korea
| | - Daehan Kim
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Hoon Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, GA, 30332, USA
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Joong Yull Park
- Department of Mechanical Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| |
Collapse
|
66
|
Ma C, Peng Y, Li H, Chen W. Organ-on-a-Chip: A New Paradigm for Drug Development. Trends Pharmacol Sci 2020; 42:119-133. [PMID: 33341248 DOI: 10.1016/j.tips.2020.11.009] [Citation(s) in RCA: 285] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/16/2023]
Abstract
The pharmaceutical industry has been desperately searching for efficient drug discovery methods. Organ-on-a-Chip, a cutting-edge technology that can emulate the physiological environment and functionality of human organs on a chip for disease modeling and drug testing, shows great potential for revolutionizing the drug development pipeline. However, successful translation of this novel engineering platform into routine pharmacological and medical scenarios remains to be realized. In this review, we discuss how the Organ-on-a-Chip technology can have critical roles in different preclinical stages of drug development and highlight the current challenges in translation and commercialization of this technology for the pharmacological and medical end-users. Moreover, this review sheds light on the future developmental trends and need for a next-generation Organ-on-a-Chip platform to bridge the gap between animal studies and clinical trials for the pharmaceutical industry.
Collapse
Affiliation(s)
- Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Yansong Peng
- Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA
| | - Hongtong Li
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY 11201, USA; Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA.
| |
Collapse
|
67
|
Hayward KL, Kouthouridis S, Zhang B. Organ-on-a-Chip Systems for Modeling Pathological Tissue Morphogenesis Associated with Fibrosis and Cancer. ACS Biomater Sci Eng 2020; 7:2900-2925. [PMID: 34275294 DOI: 10.1021/acsbiomaterials.0c01089] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tissue building does not occur exclusively during development. Even after a whole body is built from a single cell, tissue building can occur to repair and regenerate tissues of the adult body. This confers resilience and enhanced survival to multicellular organisms. However, this resiliency comes at a cost, as the potential for misdirected tissue building creates vulnerability to organ deformation and dysfunction-the hallmarks of disease. Pathological tissue morphogenesis is associated with fibrosis and cancer, which are the leading causes of morbidity and mortality worldwide. Despite being the priority of research for decades, scientific understanding of these diseases is limited and existing therapies underdeliver the desired benefits to patient outcomes. This can largely be attributed to the use of two-dimensional cell culture and animal models that insufficiently recapitulate human disease. Through the synergistic union of biological principles and engineering technology, organ-on-a-chip systems represent a powerful new approach to modeling pathological tissue morphogenesis, one with the potential to yield better insights into disease mechanisms and improved therapies that offer better patient outcomes. This Review will discuss organ-on-a-chip systems that model pathological tissue morphogenesis associated with (1) fibrosis in the context of injury-induced tissue repair and aging and (2) cancer.
Collapse
Affiliation(s)
- Kristen L Hayward
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Sonya Kouthouridis
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
68
|
Melo-Narváez MC, Stegmayr J, Wagner DE, Lehmann M. Lung regeneration: implications of the diseased niche and ageing. Eur Respir Rev 2020; 29:29/157/200222. [DOI: 10.1183/16000617.0222-2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Most chronic and acute lung diseases have no cure, leaving lung transplantation as the only option. Recent work has improved our understanding of the endogenous regenerative capacity of the lung and has helped identification of different progenitor cell populations, as well as exploration into inducing endogenous regeneration through pharmaceutical or biological therapies. Additionally, alternative approaches that aim at replacing lung progenitor cells and their progeny through cell therapy, or whole lung tissue through bioengineering approaches, have gained increasing attention. Although impressive progress has been made, efforts at regenerating functional lung tissue are still ineffective. Chronic and acute lung diseases are most prevalent in the elderly and alterations in progenitor cells with ageing, along with an increased inflammatory milieu, present major roadblocks for regeneration. Multiple cellular mechanisms, such as cellular senescence and mitochondrial dysfunction, are aberrantly regulated in the aged and diseased lung, which impairs regeneration. Existing as well as new human in vitro models are being developed, improved and adapted in order to study potential mechanisms of lung regeneration in different contexts. This review summarises recent advances in understanding endogenous as well as exogenous regeneration and the development of in vitro models for studying regenerative mechanisms.
Collapse
|
69
|
Zhang W, Huang G, Xu F. Engineering Biomaterials and Approaches for Mechanical Stretching of Cells in Three Dimensions. Front Bioeng Biotechnol 2020; 8:589590. [PMID: 33154967 PMCID: PMC7591716 DOI: 10.3389/fbioe.2020.589590] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical stretch is widely experienced by cells of different tissues in the human body and plays critical roles in regulating their behaviors. Numerous studies have been devoted to investigating the responses of cells to mechanical stretch, providing us with fruitful findings. However, these findings have been mostly observed from two-dimensional studies and increasing evidence suggests that cells in three dimensions may behave more closely to their in vivo behaviors. While significant efforts and progresses have been made in the engineering of biomaterials and approaches for mechanical stretching of cells in three dimensions, much work remains to be done. Here, we briefly review the state-of-the-art researches in this area, with focus on discussing biomaterial considerations and stretching approaches. We envision that with the development of advanced biomaterials, actuators and microengineering technologies, more versatile and predictive three-dimensional cell stretching models would be available soon for extensive applications in such fields as mechanobiology, tissue engineering, and drug screening.
Collapse
Affiliation(s)
- Weiwei Zhang
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Guoyou Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing University, Chongqing, China
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
70
|
Mejías JC, Nelson MR, Liseth O, Roy K. A 96-well format microvascularized human lung-on-a-chip platform for microphysiological modeling of fibrotic diseases. LAB ON A CHIP 2020; 20:3601-3611. [PMID: 32990704 DOI: 10.1039/d0lc00644k] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Development of organoids and microfluidic on-chip models has enabled studies of organ-level disease pathophysiologies in vitro. However, current lung-on-a-chip platforms are primarily monolayer epithelial-endothelial co-cultures, separated by a thin membrane, lacking microvasculature-networks or interstitial-fibroblasts. Here we report the design, microfabrication, and characterization of a unique microphysiological on-chip device that recapitulates the human lung interstitium-airway interface through a 3D vascular network, and normal or diseased fibroblasts encapsulated within a fibrin-collagen hydrogel underneath an airlifted airway epithelium. By incorporating fibroblasts from donors with idiopathic pulmonary fibrosis (IPF), or healthy-donor fibroblasts treated with TGF-β1, we successfully created a fibrotic, alpha smooth muscle actin (αSMA)-positive disease phenotype which led to fibrosis-like transformation in club cells and ciliated cells in the airway. Using this device platform, we further modeled the cystic fibrosis (CF) epithelium and recruitment of neutrophils to the vascular networks. Our results suggest that this microphysiological model of the human lung could enable more pathophysiologically relevant studies of complex pulmonary diseases.
Collapse
Affiliation(s)
- Joscelyn C Mejías
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Marcus Center for Therapeutic Cell Characterization and Manufacturing (MCM3), Center for ImmunoEngineering, NSF ERC for Cell Manufacturing Technologies (CMaT), The Georgia Institute of Technology, EBB 3018, 950 Atlantic Dr, Atlanta, GA 30332, USA.
| | - Michael R Nelson
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Marcus Center for Therapeutic Cell Characterization and Manufacturing (MCM3), Center for ImmunoEngineering, NSF ERC for Cell Manufacturing Technologies (CMaT), The Georgia Institute of Technology, EBB 3018, 950 Atlantic Dr, Atlanta, GA 30332, USA.
| | - Olivia Liseth
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Marcus Center for Therapeutic Cell Characterization and Manufacturing (MCM3), Center for ImmunoEngineering, NSF ERC for Cell Manufacturing Technologies (CMaT), The Georgia Institute of Technology, EBB 3018, 950 Atlantic Dr, Atlanta, GA 30332, USA.
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Marcus Center for Therapeutic Cell Characterization and Manufacturing (MCM3), Center for ImmunoEngineering, NSF ERC for Cell Manufacturing Technologies (CMaT), The Georgia Institute of Technology, EBB 3018, 950 Atlantic Dr, Atlanta, GA 30332, USA.
| |
Collapse
|
71
|
Al-Hilal TA, Keshavarz A, Kadry H, Lahooti B, Al-Obaida A, Ding Z, Li W, Kamm R, McMurtry IF, Lahm T, Nozik-Grayck E, Stenmark KR, Ahsan F. Pulmonary-arterial-hypertension (PAH)-on-a-chip: fabrication, validation and application. LAB ON A CHIP 2020; 20:3334-3345. [PMID: 32749432 PMCID: PMC7592346 DOI: 10.1039/d0lc00605j] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Currently used animal and cellular models for pulmonary arterial hypertension (PAH) only partially recapitulate its pathophysiology in humans and are thus inadequate in reproducing the hallmarks of the disease, inconsistent in portraying the sex-disparity, and unyielding to combinatorial study designs. Here we sought to deploy the ingenuity of microengineering in developing and validating a tissue chip model for human PAH. We designed and fabricated a microfluidic device to emulate the luminal, intimal, medial, adventitial, and perivascular layers of a pulmonary artery. By growing three types of pulmonary arterial cells (PACs)-endothelial, smooth muscle, and adventitial cells, we recreated the PAH pathophysiology on the device. Diseased (PAH) PACs, when grown on the chips, moved of out their designated layers and created phenomena similar to the major pathologies of human PAH: intimal thickening, muscularization, and arterial remodeling and show an endothelial to mesenchymal transition. Flow-induced stress caused control cells, grown on the chips, to undergo morphological changes and elicit arterial remodeling. Our data also suggest that the newly developed chips can be used to elucidate the sex disparity in PAH and to study the therapeutic efficacy of existing and investigational anti-PAH drugs. We believe this miniaturized device can be deployed for testing various prevailing and new hypotheses regarding the pathobiology and drug therapy in human PAH.
Collapse
Affiliation(s)
- Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 1300 Coulter Dr., Amarillo, 79119 Texas, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Matera DL, DiLillo KM, Smith MR, Davidson CD, Parikh R, Said M, Wilke CA, Lombaert IM, Arnold KB, Moore BB, Baker BM. Microengineered 3D pulmonary interstitial mimetics highlight a critical role for matrix degradation in myofibroblast differentiation. SCIENCE ADVANCES 2020; 6:eabb5069. [PMID: 32917680 PMCID: PMC11206459 DOI: 10.1126/sciadv.abb5069] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/21/2020] [Indexed: 06/11/2023]
Abstract
Fibrosis, characterized by aberrant tissue scarring from activated myofibroblasts, is often untreatable. Although the extracellular matrix becomes increasingly stiff and fibrous during disease progression, how these physical cues affect myofibroblast differentiation in 3D is poorly understood. Here, we describe a multicomponent hydrogel that recapitulates the 3D fibrous structure of interstitial tissue regions where idiopathic pulmonary fibrosis (IPF) initiates. In contrast to findings on 2D hydrogels, myofibroblast differentiation in 3D was inversely correlated with hydrogel stiffness but positively correlated with matrix fibers. Using a multistep bioinformatics analysis of IPF patient transcriptomes and in vitro pharmacologic screening, we identify matrix metalloproteinase activity to be essential for 3D but not 2D myofibroblast differentiation. Given our observation that compliant degradable 3D matrices amply support fibrogenesis, these studies demonstrate a departure from the established relationship between stiffness and myofibroblast differentiation in 2D, and provide a new 3D model for studying fibrosis and identifying antifibrotic therapeutics.
Collapse
Affiliation(s)
- Daniel L Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarina M DiLillo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Makenzee R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Ritika Parikh
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mohammed Said
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carole A Wilke
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabelle M Lombaert
- Department of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
73
|
3D Microwell Platforms for Control of Single Cell 3D Geometry and Intracellular Organization. Cell Mol Bioeng 2020; 14:1-14. [PMID: 33643464 DOI: 10.1007/s12195-020-00646-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction Cell structure and migration is impacted by the mechanical properties and geometry of the cell adhesive environment. Most studies to date investigating the effects of 3D environments on cells have not controlled geometry at the single-cell level, making it difficult to understand the influence of 3D environmental cues on single cells. Here, we developed microwell platforms to investigate the effects of 2D vs. 3D geometries on single-cell F-actin and nuclear organization. Methods We used microfabrication techniques to fabricate three polyacrylamide platforms: 3D microwells with a 3D adhesive environment (3D/3D), 3D microwells with 2D adhesive areas at the bottom only (3D/2D), and flat 2D gels with 2D patterned adhesive areas (2D/2D). We measured geometric swelling and Young's modulus of the platforms. We then cultured C2C12 myoblasts on each platform and evaluated the effects of the engineered microenvironments on F-actin structure and nuclear shape. Results We tuned the mechanical characteristics of the microfabricated platforms by manipulating the gel formulation. Crosslinker ratio strongly influenced geometric swelling whereas total polymer content primarily affected Young's modulus. When comparing cells in these platforms, we found significant effects on F-actin and nuclear structures. Our analysis showed that a 3D/3D environment was necessary to increase actin and nuclear height. A 3D/2D environment was sufficient to increase actin alignment and nuclear aspect ratio compared to a 2D/2D environment. Conclusions Using our novel polyacrylamide platforms, we were able to decouple the effects of 3D confinement and adhesive environment, finding that both influenced actin and nuclear structure.
Collapse
|
74
|
Henslee EA. Review: Dielectrophoresis in cell characterization. Electrophoresis 2020; 41:1915-1930. [DOI: 10.1002/elps.202000034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/31/2020] [Accepted: 07/14/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Erin A. Henslee
- Department of Engineering Wake Forest University 455 Vine St. Winston‐Salem USA
| |
Collapse
|
75
|
Miao K, Pan T, Mou Y, Zhang L, Xiong W, Xu Y, Yu J, Wang Y. Scutellarein inhibits BLM-mediated pulmonary fibrosis by affecting fibroblast differentiation, proliferation, and apoptosis. Ther Adv Chronic Dis 2020; 11:2040622320940185. [PMID: 32843954 PMCID: PMC7418478 DOI: 10.1177/2040622320940185] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial pulmonary disease that has a poor prognosis. Scutellarein, which is extracted from the traditional Chinese medicine Erigeron breviscapus, is used to treat a variety of diseases; however, the use of scutellarein for the treatment of pulmonary fibrosis and the related mechanisms of action have not been fully explored. Methods This study was conducted using a well-established mouse model of pulmonary fibrosis induced by bleomycin (BLM). The antifibrotic effects of scutellarein on histopathologic manifestations and fibrotic marker expression levels were examined. The effects of scutellarein on fibroblast differentiation, proliferation, and apoptosis and on related signaling pathways were next investigated to demonstrate the underlying mechanisms. Results In the present study, we found that scutellarein alleviated BLM-induced pulmonary fibrosis, as indicated by histopathologic manifestations and the expression levels of fibrotic markers. Further data demonstrated that the ability of fibroblasts to differentiate into myofibroblasts was attenuated in scutellarein-treated mice model. In addition, we obtained in vitro evidence that scutellarein inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-β/Smad signaling, inhibited cellular proliferation by repressing PI3K/Akt signaling, and increased apoptosis of fibroblasts by affecting Bax/Bcl2 signaling. Discussion In general, scutellarein might exert therapeutic effects on pulmonary fibrosis by altering the differentiation, proliferation, and apoptosis of fibroblasts. Although scutellarein has been demonstrated to be safe in mice, further studies are required to investigate the efficacy of scutellarein in patients with IPF.
Collapse
Affiliation(s)
- Kang Miao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China
| | - Ting Pan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China
| | - Yong Mou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China
| | - Lei Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China Department of Respiratory Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Wuhan Clinical Medical Research Center for Chronic Airway Diseases, Wuhan, China
| | - Jun Yu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | | |
Collapse
|
76
|
Pakshir P, Noskovicova N, Lodyga M, Son DO, Schuster R, Goodwin A, Karvonen H, Hinz B. The myofibroblast at a glance. J Cell Sci 2020; 133:133/13/jcs227900. [DOI: 10.1242/jcs.227900] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
In 1971, Gabbiani and co-workers discovered and characterized the “modification of fibroblasts into cells which are capable of an active spasm” (contraction) in rat wound granulation tissue and, accordingly, named these cells ‘myofibroblasts’. Now, myofibroblasts are not only recognized for their physiological role in tissue repair but also as cells that are key in promoting the development of fibrosis in all organs. In this Cell Science at a Glance and the accompanying poster, we provide an overview of the current understanding of central aspects of myofibroblast biology, such as their definition, activation from different precursors, the involved signaling pathways and most widely used models to study their function. Myofibroblasts will be placed into context with their extracellular matrix and with other cell types communicating in the fibrotic environment. Furthermore, the challenges and strategies to target myofibroblasts in anti-fibrotic therapies are summarized to emphasize their crucial role in disease progression.
Collapse
Affiliation(s)
- Pardis Pakshir
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Nina Noskovicova
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Monika Lodyga
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Dong Ok Son
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - Amanda Goodwin
- Nottingham NIHR Respiratory Biomedical Research Unit, University of Nottingham, Nottingham NG7 2UH, UK
| | - Henna Karvonen
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
- Respiratory Medicine, Research Unit of Internal Medicine, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, POB 20, 90029 Oulu, Finland
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
77
|
Guenat OT, Geiser T, Berthiaume F. Clinically Relevant Tissue Scale Responses as New Readouts from Organs-on-a-Chip for Precision Medicine. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:111-133. [PMID: 31961712 DOI: 10.1146/annurev-anchem-061318-114919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Organs-on-chips (OOC) are widely seen as being the next generation in vitro models able to accurately recreate the biochemical-physical cues of the cellular microenvironment found in vivo. In addition, they make it possible to examine tissue-scale functional properties of multicellular systems dynamically and in a highly controlled manner. Here we summarize some of the most remarkable examples of OOC technology's ability to extract clinically relevant tissue-level information. The review is organized around the types of OOC outputs that can be measured from the cultured tissues and transferred to clinically meaningful information. First, the creation of functional tissues-on-chip is discussed, followed by the presentation of tissue-level readouts specific to OOC, such as morphological changes, vessel formation and function, tissue properties, and metabolic functions. In each case, the clinical relevance of the extracted information is highlighted.
Collapse
Affiliation(s)
- Olivier T Guenat
- ARTORG Center for Biomedical Engineering Research, Medical Faculty, University of Bern, CH-3008 Bern, Switzerland;
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
- Thoracic Surgery Department, University Hospital of Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital and University of Bern, CH-3008 Bern, Switzerland
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
78
|
Liang W, Yang X, Wang J, Wang Y, Yang W, Liu L. Determination of Dielectric Properties of Cells using AC Electrokinetic-based Microfluidic Platform: A Review of Recent Advances. MICROMACHINES 2020; 11:E513. [PMID: 32438680 PMCID: PMC7281274 DOI: 10.3390/mi11050513] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022]
Abstract
Cell dielectric properties, a type of intrinsic property of cells, can be used as electrophysiological biomarkers that offer a label-free way to characterize cell phenotypes and states, purify clinical samples, and identify target cancer cells. Here, we present a review of the determination of cell dielectric properties using alternating current (AC) electrokinetic-based microfluidic mechanisms, including electro-rotation (ROT) and dielectrophoresis (DEP). The review covers theoretically how ROT and DEP work to extract cell dielectric properties. We also dive into the details of differently structured ROT chips, followed by a discussion on the determination of cell dielectric properties and the use of these properties in bio-related applications. Additionally, the review offers a look at the future challenges facing the AC electrokinetic-based microfluidic platform in terms of acquiring cell dielectric parameters. Our conclusion is that this platform will bring biomedical and bioengineering sciences to the next level and ultimately achieve the shift from lab-oriented research to real-world applications.
Collapse
Affiliation(s)
- Wenfeng Liang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Xieliu Yang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Junhai Wang
- School of Mechanical Engineering, Shenyang Jianzhu University, Shenyang 110168, China; (X.Y.); (J.W.)
| | - Yuechao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China;
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China;
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang 110016, China;
| |
Collapse
|
79
|
Davidson MD, Burdick JA, Wells RG. Engineered Biomaterial Platforms to Study Fibrosis. Adv Healthc Mater 2020; 9:e1901682. [PMID: 32181987 PMCID: PMC7274888 DOI: 10.1002/adhm.201901682] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/13/2022]
Abstract
Many pathologic conditions lead to the development of tissue scarring and fibrosis, which are characterized by the accumulation of abnormal extracellular matrix (ECM) and changes in tissue mechanical properties. Cells within fibrotic tissues are exposed to dynamic microenvironments that may promote or prolong fibrosis, which makes it difficult to treat. Biomaterials have proved indispensable to better understand how cells sense their extracellular environment and are now being employed to study fibrosis in many tissues. As mechanical testing of tissues becomes more routine and biomaterial tools become more advanced, the impact of biophysical factors in fibrosis are beginning to be understood. Herein, fibrosis from a materials perspective is reviewed, including the role and mechanical properties of ECM components, the spatiotemporal mechanical changes that occur during fibrosis, current biomaterial systems to study fibrosis, and emerging biomaterial systems and tools that can further the understanding of fibrosis initiation and progression. This review concludes by highlighting considerations in promoting wide-spread use of biomaterials for fibrosis investigations and by suggesting future in vivo studies that it is hoped will inspire the development of even more advanced biomaterial systems.
Collapse
Affiliation(s)
- Matthew D Davidson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rebecca G Wells
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
80
|
Loh SH, Chang CY, Huang SF, Chao SC, Lin WK, Huang EYK, Tsai CS, Tsai YT. Functional effects of urotensin-II on intracellular pH regulators in human radial artery smooth muscle cells. Peptides 2020; 126:170236. [PMID: 31874233 DOI: 10.1016/j.peptides.2019.170236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/11/2019] [Accepted: 12/16/2019] [Indexed: 02/04/2023]
Abstract
The regulation of intracellular pH (pHi) plays a vital role in various cellular functions. We previously demonstrated that three different acid extruders, the Na+-H+ exchanger (NHE), Na+-HCO3- co-transporter (NBC) and H+-linked monocarboxylate transporter (MCT), functioned together in cultured human radial artery smooth muscle cells (HRASMCs). However, the functions of acid-loading transporters in HRASMCs remain poorly understood. Urotensin II (U-II), one of the most potent vasoconstrictors, is highly expressed in many cardiovascular diseases. The aim of this present study was to determine the concentration effect of U-II (3 pM∼100 nM) on the functional activity of pHi regulators in HRASMCs. Cultured HRASMCs were derived from segments of human radial arteries obtained from patients undergoing bypass grafting. Changes in pHi recovery due to intracellular acidification and alkalization induced by NH4Cl prepulse and Na-acetate prepulse, respectively, were detected by microspectrofluorimetry with the pH-sensitive fluorescent dye BCECF. Our present study showed that (a) U-II increased the activity of NHE in a concentration-dependent manner but did not change that of NBC or MCT or resting pHi, (b) the Cl--OH- exchanger (CHE) facilitated base extrusion, and (c) U-II induced a concentration-dependent increase in the activity of CHE. In conclusion, for the first time, our results highlight a concentration-dependent increase in the activity of NHE and CHE, but not NBC and MCT, induced by U-II in HRASMCs.
Collapse
Affiliation(s)
- Shih-Hurng Loh
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan; Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan; Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Chung-Yi Chang
- Department of General Surgery, Cheng-Hsieng General Hospital, Taipei, 11200, Taiwan.
| | - Shu-Fu Huang
- Department of Pharmacy Practice, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Shih-Chi Chao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Wei-Kuo Lin
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan.
| | - Chien-Sung Tsai
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
81
|
Abstract
Systemic sclerosis (SSc) has the highest cause-specific mortality of all the connective tissue diseases, and the aetiology of this complex and heterogeneous condition remains an enigma. Current disease-modifying therapies for SSc predominantly target inflammatory and vascular pathways but have variable and unpredictable clinical efficacy, and none is curative. Moreover, many of these therapies possess undesirable safety profiles and have no appreciable effect on long-term mortality. This Review describes the most promising of the existing therapeutic targets for SSc and places them in the context of our evolving understanding of the pathophysiology of this disease. As well as taking an in-depth look at the immune, inflammatory, vascular and fibrotic pathways implicated in the pathogenesis of SSc, this Review discusses emerging treatment targets and therapeutic strategies. The article concludes with an overview of important unanswered questions in SSc research that might inform the design of future studies of treatments aimed at modifying the course of this disease.
Collapse
|
82
|
Gaspar VM, Lavrador P, Borges J, Oliveira MB, Mano JF. Advanced Bottom-Up Engineering of Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903975. [PMID: 31823448 DOI: 10.1002/adma.201903975] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/30/2019] [Indexed: 05/08/2023]
Abstract
Bottom-up tissue engineering is a promising approach for designing modular biomimetic structures that aim to recapitulate the intricate hierarchy and biofunctionality of native human tissues. In recent years, this field has seen exciting progress driven by an increasing knowledge of biological systems and their rational deconstruction into key core components. Relevant advances in the bottom-up assembly of unitary living blocks toward the creation of higher order bioarchitectures based on multicellular-rich structures or multicomponent cell-biomaterial synergies are described. An up-to-date critical overview of long-term existing and rapidly emerging technologies for integrative bottom-up tissue engineering is provided, including discussion of their practical challenges and required advances. It is envisioned that a combination of cell-biomaterial constructs with bioadaptable features and biospecific 3D designs will contribute to the development of more robust and functional humanized tissues for therapies and disease models, as well as tools for fundamental biological studies.
Collapse
Affiliation(s)
- Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Pedro Lavrador
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João Borges
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
83
|
Han HS, Lee H, You D, Nguyen VQ, Song DG, Oh BH, Shin S, Choi JS, Kim JD, Pan CH, Jo DG, Cho YW, Choi KY, Park JH. Human adipose stem cell-derived extracellular nanovesicles for treatment of chronic liver fibrosis. J Control Release 2020; 320:328-336. [PMID: 31981658 DOI: 10.1016/j.jconrel.2020.01.042] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/21/2022]
Abstract
Liver fibrosis is an excessive wound healing process that occurs in response to liver damage depending on underlying aetiologies. Currently, there are no effective therapies and FDA-approved therapeutics for the treatment of liver fibrosis except liver transplantation. Multipotent adipose-derived stem cells (ADSCs) have received significant attention as regenerative medicine for liver fibrosis owing to their advantages over stem cells with other origins. However, intrinsic limitations of stem cell therapies, such as cellular rejection and tumor formation, have impeded clinical applications of the ADSC-based liver therapeutics. To overcome these problems, the extracellular nanovesicles (ENVs) responsible for the therapeutic effect of ADSCs (A-ENVs) have shown considerable promise as cell-free therapeutics for liver diseases. However, A-ENVs have not been used for the treatment of intractable chronic liver diseases including liver fibrosis and cirrhosis. Therefore, in this study, we investigated the in vitro and in vivo antifibrotic efficacy of A-ENVs in thioacetamide-induced liver fibrosis models. A-ENVs significantly downregulated the expression of fibrogenic markers, such as matrix metalloproteinase-2, collagen-1, and alpha-smooth muscle actin. The systemic administration of A-ENVs led to high accumulation in fibrotic liver tissue and the restoration of liver functionality in liver fibrosis models through a marked reduction in α-SMA and collagen deposition. These results demonstrate the significant potential of A-ENVs for use as extracellular nanovesicles-based therapeutics in the treatment of liver fibrosis and possibly other intractable chronic liver diseases.
Collapse
Affiliation(s)
- Hwa Seung Han
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Hansang Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - DongGil You
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Quy Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dae-Geun Song
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Byeong Hoon Oh
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sol Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Ji Suk Choi
- Department of Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea; Research Institute, Exostemtech Inc., Ansan 15588, Republic of Korea
| | - Jae Dong Kim
- Research Institute, Exostemtech Inc., Ansan 15588, Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yong Woo Cho
- Department of Chemical Engineering, Hanyang University, Ansan 15588, Republic of Korea; Research Institute, Exostemtech Inc., Ansan 15588, Republic of Korea.
| | - Ki Young Choi
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea.
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
84
|
Sacchi M, Bansal R, Rouwkema J. Bioengineered 3D Models to Recapitulate Tissue Fibrosis. Trends Biotechnol 2020; 38:623-636. [PMID: 31952833 DOI: 10.1016/j.tibtech.2019.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
Abstract
Fibrosis, characterized by progressive tissue stiffening resulting in organ failure, is a growing health problem affecting millions of people worldwide. Currently, therapeutic options for tissue fibrosis are severely limited and organ transplantation is the only effective treatment for the end-stage fibrotic diseases with inherent limitations. Recent advancements in engineered 3D in vitro human disease mimic models, recapitulating the tissue pathophysiology, have provided unique state-of-the-art platforms for: (i) understanding the biological mechanisms involved in the disease pathogenesis; and (ii) high-throughput and reproducible drug screening. This review focuses on the recent multidisciplinary developments made towards advanced 3D biomimetic fibrotic tissue (liver, kidney, and lung) models that combine highly precision manufacturing techniques with high cellular functionality and biophysical (mechanical) properties.
Collapse
Affiliation(s)
- Marta Sacchi
- Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands; Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| | - Jeroen Rouwkema
- Department of Biomechanical Engineering, Technical Medical Centre, Faculty of Engineering Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
85
|
Wu H, Gu X, Li J, Wang M, Li Y, Yuan L, Wang J, Ma E. Identification of potential platelet-derived growth factor receptor α inhibitors by computational screening and binding simulations. J Mol Graph Model 2019; 96:107527. [PMID: 31918319 DOI: 10.1016/j.jmgm.2019.107527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 11/15/2022]
Abstract
Platelet-derived growth factor receptor α (PDGFRα) is considered as a promising target for the treatment of fibrotic diseases. In this study, two types of pharmacophore model, which generated by ligand-based and receptor-based method, were put forward to identify novel chemical entities as PDGFRα inhibitors. It was found that some pharmacophore characteristics established by the two approaches overlap each other. In order to elucidate detailed interactions, representative molecules were selected to predict the conformations and binding modes of the molecules by molecular docking method. The calculation results of binding free energy illustrated that the van der Waals energy and nonpolar solvation were the most prominent contribution to the interactions between the inhibitors and PDGFRα. To further verify the accuracy of the docking results and the stability of the complexes system, the binding modes of two potent PDGFRα inhibitors were examined by 100 ns molecular dynamics simulations. Herein, we reported the basic structural requirements of PDGFRα inhibitors for the first time through molecular docking and molecular dynamics simulations. Subsequently, the two pharmacophore models were used for virtual screening to query potential active molecules from Food and Drug Administration approved database. The hit molecules here might provide additional scaffolds for further optimization of PDGFRα inhibitors.
Collapse
Affiliation(s)
- Hairui Wu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Xi Gu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Jinling Li
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Mingxing Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Yanchun Li
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Lei Yuan
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| | - Enlong Ma
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, PR China.
| |
Collapse
|
86
|
Asano Y, Varga J. Rationally-based therapeutic disease modification in systemic sclerosis: Novel strategies. Semin Cell Dev Biol 2019; 101:146-160. [PMID: 31859147 DOI: 10.1016/j.semcdb.2019.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023]
Abstract
Systemic sclerosis (SSc) is a highly challenging chronic condition that is dominated by the pathogenetic triad of vascular damage, immune dysregulation/autoimmunity and fibrosis in multiple organs. A hallmark of SSc is the remarkable degree of molecular and phenotypic disease heterogeneity, which surpasses that of other complex rheumatic diseases. Disease trajectories in SSc are unpredictable and variable from patient to patient. Disease-modifying therapies for SSc are lacking, long-term morbidity is considerable and mortality remains unacceptably high. Currently-used empirical approaches to disease modification have modest and variable clinical efficacy and impact on survival, are expensive and frequently associated with unfavorable side effects, and none can be considered curative. However, research during the past several years is yielding significant advances with therapeutic potential. In particular, the application of unbiased omics-based discovery technologies to large and well-characterized SSc patient cohorts, coupled with hypothesis-testing experimental research using a variety of model systems is revealing new insights into SSc that allow formulation of a more nuanced appreciation of disease heterogeneity, and a deepening understanding of pathogenesis. Indeed, we are now presented with numerous novel and rationally-based strategies for targeted SSc therapy, several of which are currently, or expected to be shortly, undergoing clinical evaluation. In this review, we discuss promising novel therapeutic targets and rationally-based approaches to disease modification that have the potential to improve long-term outcomes in SSc.
Collapse
Affiliation(s)
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Northwestern University, Chicago, United States.
| |
Collapse
|
87
|
Abstract
Fibrosis is the abnormal deposition of extracellular matrix, which can lead to organ dysfunction, morbidity, and death. The disease burden caused by fibrosis is substantial, and there are currently no therapies that can prevent or reverse fibrosis. Metabolic alterations are increasingly recognized as an important pathogenic process that underlies fibrosis across many organ types. As a result, metabolically targeted therapies could become important strategies for fibrosis reduction. Indeed, some of the pathways targeted by antifibrotic drugs in development - such as the activation of transforming growth factor-β and the deposition of extracellular matrix - have metabolic implications. This Review summarizes the evidence to date and describes novel opportunities for the discovery and development of drugs for metabolic reprogramming, their associated challenges, and their utility in reducing fibrosis. Fibrotic therapies are potentially relevant to numerous common diseases such as cirrhosis, non-alcoholic steatohepatitis, chronic renal disease, heart failure, diabetes, idiopathic pulmonary fibrosis, and scleroderma.
Collapse
|
88
|
Asmani M, Kotei C, Hsia I, Marecki L, Wang T, Zhou C, Zhao R. Cyclic Stretching of Fibrotic Microtissue Array for Evaluation of Anti-Fibrosis Drugs. Cell Mol Bioeng 2019; 12:529-540. [PMID: 31719931 DOI: 10.1007/s12195-019-00590-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/17/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction Progression of pulmonary fibrosis, characterized by the deterioration of lung tissue's mechanical properties, is affected by respiratory motion-induced dynamic loading. Since the development of anti-fibrosis drugs faces major hurdles in animal tests and human clinical trials, preclinical models that can recapitulate fibrosis progression under physiologically-relevant cyclic loading hold great promise. However, the integration of these two functions has not been achieved in existing models. Methods Recently we developed static human lung microtissue arrays that recapitulate the progressive changes in tissue mechanics during lung fibrogenesis. In the current study, we integrate the lung microtissue array with a membrane stretching system to enable dynamic loading to the microtissues. The effects of a pro-fibrotic agent and anti-fibrosis drugs were tested under cyclic stretching. Results Cyclic stretching that mimics respiratory motion was shown to affect the cytoskeletal organization and cellular orientation in the microtissue and cause the increase in microtissue contractility and stiffness. Fibrosis induction using TGF-β1 further promoted fibrosis-related mechanical activity of the lung microtissues. Using this system, we examined the therapeutic effects of two FDA approved anti-fibrotic drugs. Our results showed that Nintedanib was able to fully inhibit TGF-β1 induced force increase but only partially inhibited stretching induced force increase. In contrast, Pirfenidone was able to fully inhibit both TGF-β1 induced force increase and stretching-induced force increase. Conclusions Together, these results highlight the pathophysiologically-relevant modeling capability of the current fibrotic microtissue system and demonstrated the potential of this system to be used for anti-fibrosis drug screening.
Collapse
Affiliation(s)
- Mohammadnabi Asmani
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Christopher Kotei
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Isaac Hsia
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Leo Marecki
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Tianjiao Wang
- Department of Industrial and Systems Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Chi Zhou
- Department of Industrial and Systems Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260 USA
| |
Collapse
|
89
|
Wang E, Rafatian N, Zhao Y, Lee A, Lai BFL, Lu RX, Jekic D, Davenport Huyer L, Knee-Walden EJ, Bhattacharya S, Backx PH, Radisic M. Biowire Model of Interstitial and Focal Cardiac Fibrosis. ACS CENTRAL SCIENCE 2019; 5:1146-1158. [PMID: 31403068 PMCID: PMC6661857 DOI: 10.1021/acscentsci.9b00052] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Indexed: 05/07/2023]
Abstract
Myocardial fibrosis is a severe global health problem due to its prevalence in all forms of cardiac diseases and direct role in causing heart failure. The discovery of efficient antifibrotic compounds has been hampered due to the lack of a physiologically relevant disease model. Herein, we present a disease model of human myocardial fibrosis and use it to establish a compound screening system. In the Biowire II platform, cardiac tissues are suspended between a pair of poly(octamethylene maleate (anhydride) citrate) (POMaC) wires. Noninvasive functional readouts are realized on the basis of the deflection of the intrinsically fluorescent polymer. The disease model is constructed to recapitulate contractile, biomechanical, and electrophysiological complexities of fibrotic myocardium. Additionally, we constructed a heteropolar integrated model with fibrotic and healthy cardiac tissues coupled together. The integrated model captures the regional heterogeneity of scar lesion, border zone, and adjacent healthy myocardium. Finally, we demonstrate the utility of the system for the evaluation of antifibrotic compounds. The high-fidelity in vitro model system combined with convenient functional readouts could potentially facilitate the development of precision medicine strategies for cardiac fibrosis modeling and establish a pipeline for preclinical compound screening.
Collapse
Affiliation(s)
- Erika
Yan Wang
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Naimeh Rafatian
- Department
of Physiology, Faculty of Medicine, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
- Toronto
General Research Institute, Toronto, Ontario M5G 2C4, Canada
| | - Yimu Zhao
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Angela Lee
- RDM
Division of Cardiovascular Medicine and Wellcome Trust Centre for
Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Benjamin Fook Lun Lai
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Rick Xingze Lu
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Danica Jekic
- McGill University, Montreal, Quebec H3A 2K6, Canada
| | - Locke Davenport Huyer
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Ericka J. Knee-Walden
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Shoumo Bhattacharya
- RDM
Division of Cardiovascular Medicine and Wellcome Trust Centre for
Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Peter H. Backx
- Department
of Physiology, Faculty of Medicine, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
- Toronto
General Research Institute, Toronto, Ontario M5G 2C4, Canada
- Department
of Biology, York University, Toronto, Ontario M3J 1P3, Canada
| | - Milica Radisic
- Institute
of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
- Toronto
General Research Institute, Toronto, Ontario M5G 2C4, Canada
- Department
of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
90
|
Abstract
Biofabrication techniques have enabled the formation of complex models of many biological tissues. We present a framework to contextualize biofabrication techniques within a disease modeling application. Fibrosis is a progressive disease interfering with tissue structure and function, which stems from an aberrant wound healing response. Epithelial injury and clot formation lead to fibroblast invasion and activation, followed by contraction and remodeling of the extracellular matrix. These stages have healthy wound healing variants in addition to the pathogenic analogs that are seen in fibrosis. This review evaluates biofabrication of a variety of phenotypic cell-based fibrosis assays. By recapitulating different contributors to fibrosis, these assays are able to evaluate biochemical pathways and therapeutic candidates for specific stages of fibrosis pathogenesis. Biofabrication of these culture models may enable phenotypic screening for improved understanding of fibrosis biology as well as improved screening of anti-fibrotic therapeutics.
Collapse
Affiliation(s)
- Cameron Yamanishi
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| | - Stephen Robinson
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| | - Shuichi Takayama
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, United States of America
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, United States of America
| |
Collapse
|
91
|
Microclot array elastometry for integrated measurement of thrombus formation and clot biomechanics under fluid shear. Nat Commun 2019; 10:2051. [PMID: 31053712 PMCID: PMC6499828 DOI: 10.1038/s41467-019-10067-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/15/2019] [Indexed: 11/08/2022] Open
Abstract
Blood clotting at the vascular injury site is a complex process that involves platelet adhesion and clot stiffening/contraction in the milieu of fluid flow. An integrated understanding of the hemodynamics and tissue mechanics regulating this process is currently lacking due to the absence of an experimental system that can simultaneously model clot formation and measure clot mechanics under shear flow. Here we develop a microfluidic-integrated microclot-array-elastometry system (clotMAT) that recapitulates dynamic changes in clot mechanics under physiological shear. Treatments with procoagulants and platelet antagonists and studies with diseased patient plasma demonstrate the ability of the system to assay clot biomechanics associated with common antiplatelet treatments and bleeding disorders. The changes of clot mechanics under biochemical treatments and shear flow demonstrate independent yet equally strong effects of these two stimulants on clot stiffening. This microtissue force sensing system may have future research and diagnostic potential for various bleeding disorders.
Collapse
|
92
|
Boudou T, Andersen T, Balland M. On the spatiotemporal regulation of cell tensional state. Exp Cell Res 2019; 378:113-117. [DOI: 10.1016/j.yexcr.2019.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/01/2019] [Accepted: 02/18/2019] [Indexed: 01/22/2023]
|
93
|
Li K, Yang X, Xue C, Zhao L, Zhang Y, Gao X. Biomimetic human lung-on-a-chip for modeling disease investigation. BIOMICROFLUIDICS 2019; 13:031501. [PMID: 31263514 PMCID: PMC6597342 DOI: 10.1063/1.5100070] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/13/2019] [Indexed: 05/11/2023]
Abstract
The lung is the primary respiratory organ of the human body and has a complicated and precise tissue structure. It comprises conductive airways formed by the trachea, bronchi and bronchioles, and many alveoli, the smallest functional units where gas-exchange occurs via the unique gas-liquid exchange interface known as the respiratory membrane. In vitro bionic simulation of the lung or its microenvironment, therefore, presents a great challenge, which requires the joint efforts of anatomy, physics, material science, cell biology, tissue engineering, and other disciplines. With the development of micromachining and miniaturization technology, the concept of a microfluidics-based organ-on-a-chip has received great attention. An organ-on-a-chip is a small cell-culture device that can accurately simulate tissue and organ functions in vitro and has the potential to replace animal models in evaluations of drug toxicity and efficacy. A lung-on-a-chip, as one of the first proposed and developed organs-on-a-chip, provides new strategies for designing a bionic lung cell microenvironment and for in vitro construction of lung disease models, and it is expected to promote the development of basic research and translational medicine in drug evaluation, toxicological detection, and disease model-building for the lung. This review summarizes current lungs-on-a-chip models based on the lung-related cellular microenvironment, including the latest advances described in studies of lung injury, inflammation, lung cancer, and pulmonary fibrosis. This model should see effective use in clinical medicine to promote the development of precision medicine and individualized diagnosis and treatment.
Collapse
Affiliation(s)
- Kaiyan Li
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Xingyuan Yang
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Chang Xue
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Lijuan Zhao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | | | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| |
Collapse
|
94
|
Li K, Yang X, Xue C, Zhao L, Zhang Y, Gao X. Biomimetic human lung-on-a-chip for modeling disease investigation. BIOMICROFLUIDICS 2019. [PMID: 31263514 DOI: 10.1063/1.5119052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The lung is the primary respiratory organ of the human body and has a complicated and precise tissue structure. It comprises conductive airways formed by the trachea, bronchi and bronchioles, and many alveoli, the smallest functional units where gas-exchange occurs via the unique gas-liquid exchange interface known as the respiratory membrane. In vitro bionic simulation of the lung or its microenvironment, therefore, presents a great challenge, which requires the joint efforts of anatomy, physics, material science, cell biology, tissue engineering, and other disciplines. With the development of micromachining and miniaturization technology, the concept of a microfluidics-based organ-on-a-chip has received great attention. An organ-on-a-chip is a small cell-culture device that can accurately simulate tissue and organ functions in vitro and has the potential to replace animal models in evaluations of drug toxicity and efficacy. A lung-on-a-chip, as one of the first proposed and developed organs-on-a-chip, provides new strategies for designing a bionic lung cell microenvironment and for in vitro construction of lung disease models, and it is expected to promote the development of basic research and translational medicine in drug evaluation, toxicological detection, and disease model-building for the lung. This review summarizes current lungs-on-a-chip models based on the lung-related cellular microenvironment, including the latest advances described in studies of lung injury, inflammation, lung cancer, and pulmonary fibrosis. This model should see effective use in clinical medicine to promote the development of precision medicine and individualized diagnosis and treatment.
Collapse
Affiliation(s)
- Kaiyan Li
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Xingyuan Yang
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Chang Xue
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Lijuan Zhao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | | | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| |
Collapse
|
95
|
Sundarakrishnan A, Zukas H, Coburn J, Bertini BT, Liu Z, Georgakoudi I, Baugh L, Dasgupta Q, Black LD, Kaplan DL. Bioengineered in Vitro Tissue Model of Fibroblast Activation for Modeling Pulmonary Fibrosis. ACS Biomater Sci Eng 2019; 5:2417-2429. [PMID: 33405750 DOI: 10.1021/acsbiomaterials.8b01262] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a complex disease of unknown etiology with no current curative treatment. Modeling pulmonary fibrotic (PF) tissue has the potential to improve our understanding of IPF disease progression and treatment. Rodent animal models do not replicate human fibroblastic foci (Hum-FF) pathology, and current iterations of in vitro model systems (e.g., collagen hydrogels, polyacrylamide hydrogels, and fibrosis-on-chip systems) are unable to replicate the three-dimensional (3D) complexity and biochemical composition of human PF tissue. Herein, we fabricated a 3D bioengineered pulmonary fibrotic (Eng-PF) tissue utilizing cell laden silk collagen type I dityrosine cross-linked hydrogels and Flexcell bioreactors. We show that silk collagen type I hydrogels have superior stability and mechanical tunability compared to other hydrogel systems. Using customized Flexcell bioreactors, we reproduced Hum-FF-like pathology with airway epithelial and microvascular endothelial cells. Eng-PF tissues can model myofibroblast differentiation and permit evaluation of antifibrotic drug treatments. Further, Eng-PF tissues could be used to model different facets of IPF disease, including epithelial injury with the addition of bleomycin and cellular recruitment by perfusion of cells through the hydrogel microchannel.
Collapse
Affiliation(s)
- Aswin Sundarakrishnan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Heather Zukas
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Jeannine Coburn
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Worcester, Massachusetts 01605, United States
| | - Brian T Bertini
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Zhiyi Liu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, Massachusetts 02114, United States
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Lauren Baugh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Queeny Dasgupta
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Department of Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, Massachusetts 02111, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
96
|
Elhebeary M, Emon MAB, Aydin O, Saif MTA. A novel technique for in situ uniaxial tests of self-assembled soft biomaterials. LAB ON A CHIP 2019; 19:1153-1161. [PMID: 30776038 PMCID: PMC6437030 DOI: 10.1039/c8lc01273c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
We introduce a novel method to form 3D biomimetic tissues from a droplet of a cell-extracellular matrix (ECM) mixture on a sensor stage and to quantify tissue force and stiffness as a function of time under optical microscopes. This method exploits advances in micro-nano fabrication and capillarity for self-assembly and self-alignment of tissues on the stage. It allows simultaneous investigation of the microstructure of the tissue in situ while its mechanical response is quantified, thus linking tissue biophysics with physiology and revealing structural-functional properties of 3D tissues. We demonstrate the functionality of the stage by studying the mechanical behavior of different cell-collagen mixtures under mechanical, chemical and electrical stimulation. This includes force evolution in cell-free collagen during curing, myotubes differentiated from muscle cell-collagen/Matrigel ECM subjected to electrical stimulation, and fibroblast-collagen tissue subjected to cancer cell conditioned media (CM) and a Rho-kinase inhibitor, Y27632. Muscle contraction decreases with increasing frequency of electrical stimulation, and fibroblasts respond to CM by increasing contractility for a short time and completely relax in the presence of Y27632 but restore force with Y27632 washout.
Collapse
Affiliation(s)
- Mohamed Elhebeary
- University of Illinois at Urbana-Champaign, 1206 W. Green St, Urbana, IL 61801, USA.
| | | | | | | |
Collapse
|
97
|
Chen Z, Zhao R. Engineered Tissue Development in Biofabricated 3D Geometrical Confinement–A Review. ACS Biomater Sci Eng 2019; 5:3688-3702. [DOI: 10.1021/acsbiomaterials.8b01195] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Zhaowei Chen
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| |
Collapse
|
98
|
Chen DQ, Feng YL, Cao G, Zhao YY. Natural Products as a Source for Antifibrosis Therapy. Trends Pharmacol Sci 2018; 39:937-952. [PMID: 30268571 DOI: 10.1016/j.tips.2018.09.002] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/29/2018] [Accepted: 09/04/2018] [Indexed: 01/15/2023]
Abstract
Although fibrosis is a final pathological feature of many chronic diseases, few interventions are available that specifically target the pathogenesis of fibrosis. Natural products are becoming increasingly recognized as effective therapies for fibrosis. The highlights of common cellular and molecular mechanisms of fibrosis facilitate the discovery of effective antifibrotic drugs. We describe some new profibrotic mechanisms and corresponding therapeutic targets using natural products. Interleukin, ephrin-B2, Gas6/TAM, Wnt/β-catenin, hedgehog pathway, PPARγ, lysophosphatidic acid, and CTGF are promising therapeutic targets. Natural products can target these mediators and inhibit chronic inflammation, myofibroblast activation, epithelial-mesenchymal transition, and extracellular matrix accumulation to alleviate fibrosis. Of note, natural products have the potential to inhibit fibrosis in one organ, simultaneously targeting fibrosis in multiple other organs, which provides us new strategies to find antifibrotic drugs.
Collapse
Affiliation(s)
- Dan-Qian Chen
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Ya-Long Feng
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Ying-Yong Zhao
- School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|