1
|
Yates AK, Murray HN, Lippmann ES. Design and optimization of a fluid flow splitting device for low-flow applications. SLAS Technol 2025; 32:100305. [PMID: 40354899 DOI: 10.1016/j.slast.2025.100305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/18/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Microfluidic devices are defined by the application of fluid flow to micron-scale features. Inherent to most experiments involving microfluidic devices is the need to precisely and reproducibly control fluid flow at the microliter scale, often through multiple inlet ports on a single device. While the number of fluid channels per device varies, perfusing multiple inputs requires either the use of multiple flow controllers (often syringe or peristaltic pumps) or the ability to evenly divide fluid across outlets. Towards the latter approach, while a handful of commercial systems exist for splitting fluid flow, these set-ups require significant financial investment, multiple flow control and sensing components, and restrict the user to a predetermined perfusion control system. Simple in-line splitting devices, such a manifolds or T junctions, fail to achieve flow splitting at low flow rates often used in microfluidic systems. To increase capabilities for flow-controlled experiments, we performed experimental analyses of the physical considerations governing even flow splitting under low flow, leading to the design of a microdevice (µ-Split) that can be directly inserted into existing microfluidic set-ups. The µ-Split allows for reproducible, even flow splitting from 10 uL/min to > 2.5 mL/min. By testing multiple device geometries in combination with multiphysics simulations, we identified the design and fabrication features underlying the splitting precision achieved by the µ-Split. Overall, this work provides a useful tool to simplify microfluidic experiments that require evenly divided flow streams, while minimizing the overall device footprint.
Collapse
Affiliation(s)
- Alexis K Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Heather N Murray
- Chemical and Biomolecular Engineering Department, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA; Chemical and Biomolecular Engineering Department, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Li G, He J, Shi J, Li X, Liu L, Ge X, Chen W, Jia J, Wang J, Yin M, Sakai Y, Sun W, Deng H, Pang Y. Bioprinting functional hepatocyte organoids derived from human chemically induced pluripotent stem cells to treat liver failure. Gut 2025:gutjnl-2024-333885. [PMID: 40032498 DOI: 10.1136/gutjnl-2024-333885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND To treat liver failure, three-dimensional (3D) bioprinting is a promising technology used to construct hepatic tissue models. However, current research on bioprinting of hepatic tissue models primarily relies on conventional single-cell-based bioprinting, where individual functional hepatocytes are dispersed and isolated within hydrogels, leading to insufficient treatment outcomes due to inadequate cell functionality. OBJECTIVE Here, we aim to bioprint a hepatic tissue model using functional hepatocyte organoids (HOs) and evaluate its liver-specific functions in vitro and in vivo. DESIGN Human chemically induced pluripotent stem cells (hCiPSCs) were used as a robust and non-genome-integrative cell source to produce highly viable and functional HOs (hCiPSC-HOs). An oxygen-permeable microwell device was used to enhance oxygen supply, ensuring high cell viability and promoting hCiPSC-HOs maturation. To maintain the long-term biofunction of hCiPSC-HOs, spheroid-based bioprinting was employed to construct hepatic tissue models (3DP-HOs). 3DP-HOs were intraperitoneally implanted in mice with liver failure. RESULTS 3DP-HOs demonstrated enhanced cell viability when compared with a model fabricated using single-cell-based bioprinting and exhibited gene profiles closely resembling hCiPSC-HOs while maintaining liver-specific functionality. Moreover, 3DP-HOs implantation significantly improved survival in mice with CCl4-induced acute-on-chronic liver failure and also Fah-/- mice with liver failure. 3DP-HOs significantly reduced liver injury, inflammation and fibrosis indices while promoting liver regeneration and biofunction expression. CONCLUSION Our bioprinted hepatic tissue model exhibits remarkable therapeutic efficacy for liver failure and holds great potential for clinical research in the field of liver regenerative medicine.
Collapse
Affiliation(s)
- Guangya Li
- Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China
| | - Jianyu He
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, People's Republic of China
| | - Jihang Shi
- Department of Gastroenterology, The Second Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xinyi Li
- International Cancer Institute, Peking University Health Science Center, Beijing, People's Republic of China
| | - Lulu Liu
- Peking University-Tsinghua University-National Institute of Biological Science Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Beijing, People's Republic of China
| | - Xinlan Ge
- Department of Hepatobiliary Surgery, Chinese PLA General Hospital, PLA, Beijing, People's Republic of China
| | - Wenhan Chen
- Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China
| | - Jun Jia
- Beijing Changping Laboratory, Beijing, People's Republic of China
| | - Jinlin Wang
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, People's Republic of China
| | - Ming Yin
- Beijing Vitalstar Biotechnology, Beijing, People's Republic of China
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, University of Tokyo, Tokyo, Japan
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, People's Republic of China
- Department of Mechanical Engineering and Mechanics, College of Engineering, Drexel University, Philadelphia, Pennsylvania, USA
| | - Hongkui Deng
- Ministry of Education (MOE) Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China
- Beijing Changping Laboratory, Beijing, People's Republic of China
- MOE Engineering Research Center of Regenerative Medicine, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, People's Republic of China
| | - Yuan Pang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Tsinghua University, Beijing, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Beijing, People's Republic of China
| |
Collapse
|
3
|
Li X, Wang M, Davis TP, Zhang L, Qiao R. Advancing Tissue Culture with Light-Driven 3D-Printed Microfluidic Devices. BIOSENSORS 2024; 14:301. [PMID: 38920605 PMCID: PMC11201418 DOI: 10.3390/bios14060301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Three-dimensional (3D) printing presents a compelling alternative for fabricating microfluidic devices, circumventing certain limitations associated with traditional soft lithography methods. Microfluidics play a crucial role in the biomedical sciences, particularly in the creation of tissue spheroids and pharmaceutical research. Among the various 3D printing techniques, light-driven methods such as stereolithography (SLA), digital light processing (DLP), and photopolymer inkjet printing have gained prominence in microfluidics due to their rapid prototyping capabilities, high-resolution printing, and low processing temperatures. This review offers a comprehensive overview of light-driven 3D printing techniques used in the fabrication of advanced microfluidic devices. It explores biomedical applications for 3D-printed microfluidics and provides insights into their potential impact and functionality within the biomedical field. We further summarize three light-driven 3D printing strategies for producing biomedical microfluidic systems: direct construction of microfluidic devices for cell culture, PDMS-based microfluidic devices for tissue engineering, and a modular SLA-printed microfluidic chip to co-culture and monitor cells.
Collapse
Affiliation(s)
| | | | | | - Liwen Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ruirui Qiao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
4
|
Ponmozhi J, Dhinakaran S, Kocsis D, Iván K, Erdő F. Models for barrier understanding in health and disease in lab-on-a-chips. Tissue Barriers 2024; 12:2221632. [PMID: 37294075 PMCID: PMC11042069 DOI: 10.1080/21688370.2023.2221632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
The maintenance of body homeostasis relies heavily on physiological barriers. Dysfunction of these barriers can lead to various pathological processes, including increased exposure to toxic materials and microorganisms. Various methods exist to investigate barrier function in vivo and in vitro. To investigate barrier function in a highly reproducible manner, ethically, and high throughput, researchers have turned to non-animal techniques and micro-scale technologies. In this comprehensive review, the authors summarize the current applications of organ-on-a-chip microfluidic devices in the study of physiological barriers. The review covers the blood-brain barrier, ocular barriers, dermal barrier, respiratory barriers, intestinal, hepatobiliary, and renal/bladder barriers under both healthy and pathological conditions. The article then briefly presents placental/vaginal, and tumour/multi-organ barriers in organ-on-a-chip devices. Finally, the review discusses Computational Fluid Dynamics in microfluidic systems that integrate biological barriers. This article provides a concise yet informative overview of the current state-of-the-art in barrier studies using microfluidic devices.
Collapse
Affiliation(s)
- J. Ponmozhi
- Microfluidics Laboratory, Department of Mechanical Engineering, IPS Academy-Institute of Engineering Science, Indore, India
| | - S. Dhinakaran
- The Centre for Fluid Dynamics, Department of Mechanical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Kristóf Iván
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
5
|
Milton LA, Viglione MS, Ong LJY, Nordin GP, Toh YC. Vat photopolymerization 3D printed microfluidic devices for organ-on-a-chip applications. LAB ON A CHIP 2023; 23:3537-3560. [PMID: 37476860 PMCID: PMC10448871 DOI: 10.1039/d3lc00094j] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Organs-on-a-chip, or OoCs, are microfluidic tissue culture devices with micro-scaled architectures that repeatedly achieve biomimicry of biological phenomena. They are well positioned to become the primary pre-clinical testing modality as they possess high translational value. Current methods of fabrication have facilitated the development of many custom OoCs that have generated promising results. However, the reliance on microfabrication and soft lithographic fabrication techniques has limited their prototyping turnover rate and scalability. Additive manufacturing, known commonly as 3D printing, shows promise to expedite this prototyping process, while also making fabrication easier and more reproducible. We briefly introduce common 3D printing modalities before identifying two sub-types of vat photopolymerization - stereolithography (SLA) and digital light processing (DLP) - as the most advantageous fabrication methods for the future of OoC development. We then outline the motivations for shifting to 3D printing, the requirements for 3D printed OoCs to be competitive with the current state of the art, and several considerations for achieving successful 3D printed OoC devices touching on design and fabrication techniques, including a survey of commercial and custom 3D printers and resins. In all, we aim to form a guide for the end-user to facilitate the in-house generation of 3D printed OoCs, along with the future translation of these important devices.
Collapse
Affiliation(s)
- Laura A Milton
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
| | - Matthew S Viglione
- Department of Electrical and Computer Engineering, Brigham Young University, Provo, Utah, USA.
| | - Louis Jun Ye Ong
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
| | - Gregory P Nordin
- Department of Electrical and Computer Engineering, Brigham Young University, Provo, Utah, USA.
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane, Australia
- Centre for Microbiome Research, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
6
|
A Cataño J, Farthing S, Mascarenhas Z, Lake N, Yarlagadda PKDV, Li Z, Toh YC. A User-Centric 3D-Printed Modular Peristaltic Pump for Microfluidic Perfusion Applications. MICROMACHINES 2023; 14:mi14050930. [PMID: 37241553 DOI: 10.3390/mi14050930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023]
Abstract
Microfluidic organ-on-a-chip (OoC) technology has enabled studies on dynamic physiological conditions as well as being deployed in drug testing applications. A microfluidic pump is an essential component to perform perfusion cell culture in OoC devices. However, it is challenging to have a single pump that can fulfil both the customization function needed to mimic a myriad of physiological flow rates and profiles found in vivo and multiplexing requirements (i.e., low cost, small footprint) for drug testing operations. The advent of 3D printing technology and open-source programmable electronic controllers presents an opportunity to democratize the fabrication of mini-peristaltic pumps suitable for microfluidic applications at a fraction of the cost of commercial microfluidic pumps. However, existing 3D-printed peristaltic pumps have mainly focused on demonstrating the feasibility of using 3D printing to fabricate the structural components of the pump and neglected user experience and customization capability. Here, we present a user-centric programmable 3D-printed mini-peristaltic pump with a compact design and low manufacturing cost (~USD 175) suitable for perfusion OoC culture applications. The pump consists of a user-friendly, wired electronic module that controls the operation of a peristaltic pump module. The peristaltic pump module comprises an air-sealed stepper motor connected to a 3D-printed peristaltic assembly, which can withstand the high-humidity environment of a cell culture incubator. We demonstrated that this pump allows users to either program the electronic module or use different-sized tubing to deliver a wide range of flow rates and flow profiles. The pump also has multiplexing capability as it can accommodate multiple tubing. The performance and user-friendliness of this low-cost, compact pump can be easily deployed for various OoC applications.
Collapse
Affiliation(s)
- Jorge A Cataño
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
| | - Steven Farthing
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Zeus Mascarenhas
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Nathaniel Lake
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
| | - Prasad K D V Yarlagadda
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
- School of Engineering, University of Southern Queensland, Springfield Central 4300, Australia
| | - Zhiyong Li
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane 4000, Australia
- Centre for Biomedical Technologies, Queensland University of Technology, Kelvin Grove 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Kelvin Grove 4059, Australia
- Centre for Microbiome Research, Queensland University of Technology, Woolloongabba 4102, Australia
| |
Collapse
|
7
|
Ong LJY, Fan X, Rujia Sun A, Mei L, Toh YC, Prasadam I. Controlling Microenvironments with Organs-on-Chips for Osteoarthritis Modelling. Cells 2023; 12:579. [PMID: 36831245 PMCID: PMC9954502 DOI: 10.3390/cells12040579] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Osteoarthritis (OA) remains a prevalent disease affecting more than 20% of the global population, resulting in morbidity and lower quality of life for patients. The study of OA pathophysiology remains predominantly in animal models due to the complexities of mimicking the physiological environment surrounding the joint tissue. Recent development in microfluidic organ-on-chip (OoC) systems have demonstrated various techniques to mimic and modulate tissue physiological environments. Adaptations of these techniques have demonstrated success in capturing a joint tissue's tissue physiology for studying the mechanism of OA. Adapting these techniques and strategies can help create human-specific in vitro models that recapitulate the cellular processes involved in OA. This review aims to comprehensively summarise various demonstrations of microfluidic platforms in mimicking joint microenvironments for future platform design iterations.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| | - Xiwei Fan
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Antonia Rujia Sun
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Lin Mei
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
- Max Planck Queensland Centre (MPQC) for the Materials Science of Extracellular Matrices, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Centre for Microbiome Research, Queensland University of Technology, Brisbane City, QLD 4000, Australia
| | - Indira Prasadam
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- Center for Biomedical Technologies, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
8
|
Makkar H, Zhou Y, Tan KS, Lim CT, Sriram G. Modeling Crevicular Fluid Flow and Host-Oral Microbiome Interactions in a Gingival Crevice-on-Chip. Adv Healthc Mater 2023; 12:e2202376. [PMID: 36398428 DOI: 10.1002/adhm.202202376] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/07/2022] [Indexed: 11/21/2022]
Abstract
Gingival crevice and gingival crevicular fluid (GCF) flow play a crucial role at the gingiva-oral microbiome interface which contributes toward maintaining the balance between gingival health and periodontal disease. Interstitial flow of GCF strongly impacts the host-microbiome interactions and tissue responses. However, currently available in vitro preclinical models largely disregard the dynamic nature of gingival crevicular microenvironment, thus limiting the progress in the development of periodontal therapeutics. Here, a proof-of-principle "gingival crevice-on-chip" microfluidic platform to culture gingival connective tissue equivalent (CTE) under dynamic interstitial fluid flow mimicking the GCF is described. On-chip co-culture using oral symbiont (Streptococcus oralis) shows the potential to recapitulate microbial colonization, formation of biofilm-like structures at the tissue-microbiome interface, long-term co-culture, and bacterial clearance secondary to simulated GCF (s-GCF) flow. Further, on-chip exposure of the gingival CTEs to the toll-like receptor-2 (TLR-2) agonist or periodontal pathogen Fusobacterium nucleatum demonstrates the potential to mimic early gingival inflammation. In contrast to direct exposure, the induction of s-GCF flow toward the bacterial front attenuates the secretion of inflammatory mediators demonstrating the protective effect of GCF flow. This proposed in vitro platform offers the potential to study complex host-microbe interactions in periodontal disease and the development of periodontal therapeutics under near-microphysiological conditions.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
| | - Ying Zhou
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, Singapore
| | - Kai Soo Tan
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore.,ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore, 119085, Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore.,Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore.,ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore, 119085, Singapore
| |
Collapse
|
9
|
Yun C, Kim SH, Jung YS. Current Research Trends in the Application of In Vitro Three-Dimensional Models of Liver Cells. Pharmaceutics 2022; 15:pharmaceutics15010054. [PMID: 36678683 PMCID: PMC9866911 DOI: 10.3390/pharmaceutics15010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The liver produces and stores various nutrients that are necessary for the body and serves as a chemical plant, metabolizing carbohydrates, fats, hormones, vitamins, and minerals. It is also a vital organ for detoxifying drugs and exogenous harmful substances. Culturing liver cells in vitro under three-dimensional (3D) conditions is considered a primary mechanism for liver tissue engineering. The 3D cell culture system is designed to allow cells to interact in an artificially created environment and has the advantage of mimicking the physiological characteristics of cells in vivo. This system facilitates contact between the cells and the extracellular matrix. Several technically different approaches have been proposed, including bioreactors, chips, and plate-based systems in fluid or static media composed of chemically diverse materials. Compared to conventional two-dimensional monolayer culture in vitro models, the ability to predict the function of the tissues, including the drug metabolism and chemical toxicity, has been enhanced by developing three-dimensional liver culture models. This review discussed the methodology of 3D cell cultures and summarized the advantages of an in vitro liver platform using 3D culture technology.
Collapse
|
10
|
Ishida-Ishihara S, Takada R, Furusawa K, Ishihara S, Haga H. Improvement of the cell viability of hepatocytes cultured in three-dimensional collagen gels using pump-free perfusion driven by water level difference. Sci Rep 2022; 12:20269. [PMID: 36434099 PMCID: PMC9700666 DOI: 10.1038/s41598-022-24423-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Cell-containing collagen gels are one of the materials employed in tissue engineering and drug testing. A collagen gel is a useful three-dimensional (3D) scaffold that improves various cell functions compared to traditional two-dimensional plastic substrates. However, owing to poor nutrient availability, cells are not viable in thick collagen gels. Perfusion is an effective method for supplying nutrients to the gel. In this study, we maintained hepatocytes embedded in a 3D collagen gel using a simple pump-free perfusion cell culture system with ordinary cell culture products. Flow was generated by the difference in water level in the culture medium. Hepatocytes were found to be viable in a collagen gel of thickness 3.26 (± 0.16 S.E.)-mm for 3 days. In addition, hepatocytes had improved proliferation and gene expression related to liver function in a 3D collagen gel compared to a 2D culture dish. These findings indicate that our perfusion method is useful for investigating the cellular functions of 3D hydrogels.
Collapse
Affiliation(s)
- Sumire Ishida-Ishihara
- grid.39158.360000 0001 2173 7691Department of Functional Life Sciences, Faculty of Advanced Life Science, Hokkaido University, N21-W11, Kita-Ku, Sapporo, 001-0021 Japan
| | - Ryota Takada
- grid.39158.360000 0001 2173 7691Division of Life Science, Graduate School of Life Science, Hokkaido University, N10-W8, Kita-Ku, Sapporo, 060-0810 Japan
| | - Kazuya Furusawa
- grid.440871.e0000 0000 9829 078XFaculty of Environmental and Information Sciences, Fukui University of Technology, Gakuen 3-6-1, Fukui, 910-8505 Japan
| | - Seiichiro Ishihara
- grid.39158.360000 0001 2173 7691Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, N10-W8, Kita-Ku, Sapporo, 060-0810 Japan ,grid.39158.360000 0001 2173 7691Soft Matter GI-CoRE, Hokkaido University, N21-W11, Kita-Ku, Sapporo, 001-0021 Japan ,grid.39158.360000 0001 2173 7691Hokkaido University, Room 2-602, Science Bld., N10-W8, Kita-Ku, Sapporo, 060-0810 Japan
| | - Hisashi Haga
- grid.39158.360000 0001 2173 7691Department of Advanced Transdisciplinary Sciences, Faculty of Advanced Life Science, Hokkaido University, N10-W8, Kita-Ku, Sapporo, 060-0810 Japan ,grid.39158.360000 0001 2173 7691Soft Matter GI-CoRE, Hokkaido University, N21-W11, Kita-Ku, Sapporo, 001-0021 Japan ,grid.39158.360000 0001 2173 7691Hokkaido University, Room 2-612, Science Bld., N10-W8, Kita-Ku, Sapporo, 060-0810 Japan
| |
Collapse
|
11
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
12
|
Dalsbecker P, Beck Adiels C, Goksör M. Liver-on-a-chip devices: the pros and cons of complexity. Am J Physiol Gastrointest Liver Physiol 2022; 323:G188-G204. [PMID: 35819853 DOI: 10.1152/ajpgi.00346.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Physiologically relevant and broadly applicable liver cell culture platforms are of great importance in both drug development and disease modeling. Organ-on-a-chip systems offer a promising alternative to conventional, static two-dimensional (2-D) cultures, providing much-needed cues such as perfusion, shear stress, and three-dimensional (3-D) cell-cell communication. However, such devices cover a broad range of complexity both in manufacture and in implementation. In this review, we summarize the key features of the human liver that should be reflected in a physiologically relevant liver-on-a-chip model. We also discuss different material properties of importance in producing liver-on-a-chip devices and summarize recent and current progress in the field, highlighting different types of devices at different levels of complexity.
Collapse
Affiliation(s)
| | | | - Mattias Goksör
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
13
|
Ong LJY, Chia S, Wong SQR, Zhang X, Chua H, Loo JM, Chua WY, Chua C, Tan E, Hentze H, Tan IB, DasGupta R, Toh YC. A comparative study of tumour-on-chip models with patient-derived xenografts for predicting chemotherapy efficacy in colorectal cancer patients. Front Bioeng Biotechnol 2022; 10:952726. [PMID: 36147524 PMCID: PMC9488115 DOI: 10.3389/fbioe.2022.952726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Inter-patient and intra-tumour heterogeneity (ITH) have prompted the need for a more personalised approach to cancer therapy. Although patient-derived xenograft (PDX) models can generate drug response specific to patients, they are not sustainable in terms of cost and time and have limited scalability. Tumour Organ-on-Chip (OoC) models are in vitro alternatives that can recapitulate some aspects of the 3D tumour microenvironment and can be scaled up for drug screening. While many tumour OoC systems have been developed to date, there have been limited validation studies to ascertain whether drug responses obtained from tumour OoCs are comparable to those predicted from patient-derived xenograft (PDX) models. In this study, we established a multiplexed tumour OoC device, that consists of an 8 × 4 array (32-plex) of culture chamber coupled to a concentration gradient generator. The device enabled perfusion culture of primary PDX-derived tumour spheroids to obtain dose-dependent response of 5 distinct standard-of-care (SOC) chemotherapeutic drugs for 3 colorectal cancer (CRC) patients. The in vitro efficacies of the chemotherapeutic drugs were rank-ordered for individual patients and compared to the in vivo efficacy obtained from matched PDX models. We show that quantitative correlation analysis between the drug efficacies predicted via the microfluidic perfusion culture is predictive of response in animal PDX models. This is a first study showing a comparative framework to quantitatively correlate the drug response predictions made by a microfluidic tumour organ-on-chip (OoC) model with that of PDX animal models.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QL, Australia
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QL, Australia
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
| | - Shumei Chia
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Stephen Qi Rong Wong
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
- Biological Resource Centre, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Xiaoqian Zhang
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Huiwen Chua
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Jia Min Loo
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Wei Yong Chua
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Clarinda Chua
- National Cancer Centre Singapore, Singapore, Singapore
| | - Emile Tan
- Singapore General Hospital, Singapore, Singapore
| | - Hannes Hentze
- Experimental, Drug Development Centre, A*STAR, Singapore, Singapore
| | - Iain Beehuat Tan
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
- National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
- *Correspondence: Ramanuj DasGupta, ; Yi-Chin Toh,
| | - Yi-Chin Toh
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QL, Australia
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QL, Australia
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- *Correspondence: Ramanuj DasGupta, ; Yi-Chin Toh,
| |
Collapse
|
14
|
Singh VK, Seed TM. Acute radiation syndrome drug discovery using organ-on-chip platforms. Expert Opin Drug Discov 2022; 17:865-878. [PMID: 35838021 DOI: 10.1080/17460441.2022.2099833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION : The high attrition rate during drug development remains a challenge that costs a significant amount of time and money. Improving the probabilities of success during the early stages of radiation medical countermeasure (MCM) development for approval by the United States Food and Drug Administration (US FDA) following the Animal Rule will reduce this burden. For optimal development of MCMs, we need suitable and efficient radiation injury models with high biological relevance for evaluating drug efficacy as well as biomarker discovery and validation. AREA COVERED This article focuses on new technologies involving various organs-on-chip platforms. Of late, there have been rapid development of these technologies, especially in terms of mimicking both normal and abnormal physiological conditions. Here, we suggest possible applications of these novel systems for the discovery and development of radiation MCMs for the acute radiation syndrome (ARS). We offer preliminary information on the utility of one such system for MCM research and discovery for the ARS condition. EXPERT OPINION : Each organ-on-a-chip system has its own strengths and shortcomings. As such, the system selected for MCM discovery, development, and regulatory approval should be carefully considered and optimized to the fullest extent in order to augment successful drug testing and the minimization of attrition rates of candidate agents. The recent encouraging progress with organ-on-a-chip technology will likely lead to additional radiation MCMs for ARS approved by the US FDA. The acceptance of organ-on-a-chip technology may be a promising step toward improving the success rate of pharmaceuticals in MCM development.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD, USA
| |
Collapse
|
15
|
Chong LH, Ching T, Farm HJ, Grenci G, Chiam KH, Toh YC. Integration of a microfluidic multicellular coculture array with machine learning analysis to predict adverse cutaneous drug reactions. LAB ON A CHIP 2022; 22:1890-1904. [PMID: 35348137 DOI: 10.1039/d1lc01140e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adverse cutaneous reactions are potentially life-threatening skin side effects caused by drugs administered into the human body. The availability of a human-specific in vitro platform that can prospectively screen drugs and predict this risk is therefore of great importance to drug safety. However, since adverse cutaneous drug reactions are mediated by at least 2 distinct mechanisms, both involving systemic interactions between liver, immune and dermal tissues, existing in vitro skin models have not been able to comprehensively recapitulate these complex, multi-cellular interactions to predict the skin-sensitization potential of drugs. Here, we report a novel in vitro drug screening platform, which comprises a microfluidic multicellular coculture array (MCA) to model different mechanisms-of-action using a collection of simplistic cellular assays. The resultant readouts are then integrated with a machine-learning algorithm to predict the skin sensitizing potential of systemic drugs. The MCA consists of 4 cell culture compartments connected by diffusion microchannels to enable crosstalk between hepatocytes that generate drug metabolites, antigen-presenting cells (APCs) that detect the immunogenicity of the drug metabolites, and keratinocytes and dermal fibroblasts, which collectively determine drug metabolite-induced FasL-mediated apoptosis. A single drug screen using the MCA can simultaneously generate 5 readouts, which are integrated using support vector machine (SVM) and principal component analysis (PCA) to classify and visualize the drugs as skin sensitizers or non-skin sensitizers. The predictive performance of the MCA and SVM classification algorithm is then validated through a pilot screen of 11 drugs labelled by the US Food and Drug Administration (FDA), including 7 skin-sensitizing and 4 non-skin sensitizing drugs, using stratified 4-fold cross-validation (CV) on SVM. The predictive performance of our in vitro model achieves an average of 87.5% accuracy (correct prediction rate), 75% specificity (prediction rate of true negative drugs), and 100% sensitivity (prediction rate of true positive drugs). We then employ the MCA and the SVM training algorithm to prospectively identify the skin-sensitizing likelihood and mechanism-of-action for obeticholic acid (OCA), a farnesoid X receptor (FXR) agonist which has undergone clinical trials for non-alcoholic steatohepatitis (NASH) with well-documented cutaneous side effects.
Collapse
Affiliation(s)
- Lor Huai Chong
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Bioinformatics Institute, ASTAR, 30 Biopolis St, Singapore 138671, Singapore
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, 47500, Malaysia
| | - Terry Ching
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore 487372, Singapore
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8 Somapah Rd, Singapore 487372, Singapore
| | - Hui Jia Farm
- Department of Computer Science, University of Oxford, Oxford, OX1 3QD, UK
| | - Gianluca Grenci
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Mechanobiology Institute, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Keng-Hwee Chiam
- Bioinformatics Institute, ASTAR, 30 Biopolis St, Singapore 138671, Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- School of Mechanical Medical & Process Engineering, Queensland University of Technology, 2 George St, Brisbane, QLD 4000, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
16
|
Baddal B. Microfluidic Organ-Chips and Infectious Diseases: Insights from the Development and Applications Perspective. CYPRUS JOURNAL OF MEDICAL SCIENCES 2022. [DOI: 10.4274/cjms.2020.1426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
17
|
Cook SR, Musgrove HB, Throckmorton AL, Pompano RR. Microscale impeller pump for recirculating flow in organs-on-chip and microreactors. LAB ON A CHIP 2022; 22:605-620. [PMID: 34988560 PMCID: PMC8892988 DOI: 10.1039/d1lc01081f] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Fluid flow is an integral part of microfluidic and organ-on-chip technology, ideally providing biomimetic fluid, cell, and nutrient exchange as well as physiological or pathological shear stress. Currently, many of the pumps that actively perfuse fluid at biomimetic flow rates are incompatible with use inside cell culture incubators, require many tubing connections, or are too large to run many devices in a confined space. To address these issues, we developed a user-friendly impeller pump that uses a 3D-printed device and impeller to recirculate fluid and cells on-chip. Impeller rotation was driven by a rotating magnetic field generated by magnets mounted on a computer fan; this pump platform required no tubing connections and could accommodate up to 36 devices at once in a standard cell culture incubator. A computational model was used to predict shear stress, velocity, and changes in pressure throughout the device. The impeller pump generated biomimetic fluid velocities (50-6400 μm s-1) controllable by tuning channel and inlet dimensions and the rotational speed of the impeller, which were comparable to the order of magnitude of the velocities predicted by the computational model. Predicted shear stress was in the physiological range throughout the microchannel and over the majority of the impeller. The impeller pump successfully recirculated primary murine splenocytes for 1 h and Jurkat T cells for 24 h with no impact on cell viability, showing the impeller pump's feasibility for white blood cell recirculation on-chip. In the future, we envision that this pump will be integrated into single- or multi-tissue platforms to study communication between organs.
Collapse
Affiliation(s)
- Sophie R Cook
- Departments of Chemistry and Biomedical Engineering, University of Virginia, 248 McCormick Rd, Charlottesville, VA 22904, USA.
| | - Hannah B Musgrove
- Departments of Chemistry and Biomedical Engineering, University of Virginia, 248 McCormick Rd, Charlottesville, VA 22904, USA.
| | - Amy L Throckmorton
- BioCirc Research Laboratory, School of Biomedical Engineering, Science, and Health Systems, Philadelphia, Drexel University, Philadelphia, PA, USA
| | - Rebecca R Pompano
- Departments of Chemistry and Biomedical Engineering, University of Virginia, 248 McCormick Rd, Charlottesville, VA 22904, USA.
| |
Collapse
|
18
|
Handin N, Mickols E, Ölander M, Rudfeldt J, Blom K, Nyberg F, Senkowski W, Urdzik J, Maturi V, Fryknäs M, Artursson P. Conditions for maintenance of hepatocyte differentiation and function in 3D cultures. iScience 2021; 24:103235. [PMID: 34746700 PMCID: PMC8551077 DOI: 10.1016/j.isci.2021.103235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/02/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Spheroid cultures of primary human hepatocytes (PHH) are used in studies of hepatic drug metabolism and toxicity. The cultures are maintained under different conditions, with possible confounding results. We performed an in-depth analysis of the influence of various culture conditions to find the optimal conditions for the maintenance of an in vivo like phenotype. The formation, protein expression, and function of PHH spheroids were followed for three weeks in a high-throughput 384-well format. Medium composition affected spheroid histology, global proteome profile, drug metabolism and drug-induced toxicity. No epithelial-mesenchymal transition was observed. Media with fasting glucose and insulin levels gave spheroids with phenotypes closest to normal PHH. The most expensive medium resulted in PHH features most divergent from that of native PHH. Our results provide a protocol for culture of healthy PHH with maintained function - a prerequisite for studies of hepatocyte homeostasis and more reproducible hepatocyte research. 3D spheroid cultures were established in 384-well format Eight different media variants were used to optimize the 3D cultures Optimized William's medium was as good as expensive commercial medium The 3D cultures were used to study drug metabolism and toxicity
Collapse
Affiliation(s)
- Niklas Handin
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| | - Evgeniya Mickols
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| | - Magnus Ölander
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| | - Jakob Rudfeldt
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Kristin Blom
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Frida Nyberg
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Wojciech Senkowski
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden.,Biotech Research & Innovation Centre (BRIC) and Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Jozef Urdzik
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Varun Maturi
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| | - Mårten Fryknäs
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, 75123 Uppsala, Sweden
| |
Collapse
|
19
|
Mendoza-Martinez AK, Loessner D, Mata A, Azevedo HS. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials. Cancers (Basel) 2021; 13:5745. [PMID: 34830897 PMCID: PMC8616551 DOI: 10.3390/cancers13225745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
Collapse
Affiliation(s)
- Ana Karen Mendoza-Martinez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniela Loessner
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia;
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., 01069 Dresden, Germany
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
20
|
Aranda Hernandez J, Heuer C, Bahnemann J, Szita N. Microfluidic Devices as Process Development Tools for Cellular Therapy Manufacturing. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 179:101-127. [PMID: 34410457 DOI: 10.1007/10_2021_169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular therapies are creating a paradigm shift in the biomanufacturing industry. Particularly for autologous therapies, small-scale processing methods are better suited than the large-scale approaches that are traditionally employed in the industry. Current small-scale methods for manufacturing personalized cell therapies, however, are labour-intensive and involve a number of 'open events'. To overcome these challenges, new cell manufacturing platforms following a GMP-in-a-box concept have recently come on the market (GMP: Good Manufacturing Practice). These are closed automated systems with built-in pumps for fluid handling and sensors for in-process monitoring. At a much smaller scale, microfluidic devices exhibit many of the same features as current GMP-in-a-box systems. They are closed systems, fluids can be processed and manipulated, and sensors integrated for real-time detection of process variables. Fabricated from polymers, they can be made disposable, i.e. single-use. Furthermore, microfluidics offers exquisite spatiotemporal control over the cellular microenvironment, promising both reproducibility and control of outcomes. In this chapter, we consider the challenges in cell manufacturing, highlight recent advances of microfluidic devices for each of the main process steps, and summarize our findings on the current state of the art. As microfluidic cell culture devices have been reported for both adherent and suspension cell cultures, we report on devices for the key process steps, or unit operations, of both stem cell therapies and cell-based immunotherapies.
Collapse
Affiliation(s)
| | - Christopher Heuer
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Janina Bahnemann
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Nicolas Szita
- Biochemical Engineering Department, University College London (UCL), London, UK.
| |
Collapse
|
21
|
A versatile microfluidic tool for the 3D culture of HepaRG cells seeded at various stages of differentiation. Sci Rep 2021; 11:14075. [PMID: 34234159 DOI: 10.1038/s41598-021-92011-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/03/2021] [Indexed: 11/09/2022] Open
Abstract
The development of livers-on-a-chip aims to provide pharmaceutical companies with reliable systems to perform drug screening and toxicological studies. To that end, microfluidic systems are engineered to mimic the functions and architecture of this organ. In this context we have designed a device that reproduces series of liver microarchitectures, each permitting the 3D culture of hepatocytes by confining them to a chamber that is separated from the medium conveying channel by very thin slits. We modified the structure to ensure its compatibility with the culture of hepatocytes from different sources. Our device was adapted to the migratory and adhesion properties of the human HepaRG cell line at various stages of differentiation. Using this device, it was possible to keep the cells alive for more than 14 days, during which they achieved a 3D organisation and acquired or maintained their differentiation into hepatocytes. Albumin secretion as well as functional bile canaliculi were confirmed on the liver-on-a-chip. Finally, an acetaminophen toxicological assay was performed. With its multiple micro-chambers for hepatocyte culture, this microfluidic device architecture offers a promising opportunity to provide new tools for drug screening applications.
Collapse
|
22
|
Yazdian Kashani S, Keshavarz Moraveji M, Bonakdar S. Computational and experimental studies of a cell-imprinted-based integrated microfluidic device for biomedical applications. Sci Rep 2021; 11:12130. [PMID: 34108580 PMCID: PMC8190060 DOI: 10.1038/s41598-021-91616-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023] Open
Abstract
It has been proved that cell-imprinted substrates molded from template cells can be used for the re-culture of that cell while preserving its normal behavior or to differentiate the cultured stem cells into the template cell. In this study, a microfluidic device was presented to modify the previous irregular cell-imprinted substrate and increase imprinting efficiency by regular and objective cell culture. First, a cell-imprinted substrate from template cells was prepared using a microfluidic chip in a regular pattern. Another microfluidic chip with the same pattern was then aligned on the cell-imprinted substrate to create a chondrocyte-imprinted-based integrated microfluidic device. Computational fluid dynamics (CFD) simulations were used to obtain suitable conditions for injecting cells into the microfluidic chip before performing experimental evaluations. In this simulation, the effect of input flow rate, number per unit volume, and size of injected cells in two different chip sizes were examined on exerted shear stress and cell trajectories. This numerical simulation was first validated with experiments with cell lines. Finally, chondrocyte was used as template cell to evaluate the chondrogenic differentiation of adipose-derived mesenchymal stem cells (ADSCs) in the chondrocyte-imprinted-based integrated microfluidic device. ADSCs were positioned precisely on the chondrocyte patterns, and without using any chemical growth factor, their fibroblast-like morphology was modified to the spherical morphology of chondrocytes after 14 days of culture. Both immunostaining and gene expression analysis showed improvement in chondrogenic differentiation compared to traditional imprinting methods. This study demonstrated the effectiveness of cell-imprinted-based integrated microfluidic devices for biomedical applications.
Collapse
Affiliation(s)
- Sepideh Yazdian Kashani
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 1591634311, Iran
| | - Mostafa Keshavarz Moraveji
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 1591634311, Iran.
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran.
| |
Collapse
|
23
|
Duivenvoorde LPM, Louisse J, Pinckaers NET, Nguyen T, van der Zande M. Comparison of gene expression and biotransformation activity of HepaRG cells under static and dynamic culture conditions. Sci Rep 2021; 11:10327. [PMID: 33990636 PMCID: PMC8121841 DOI: 10.1038/s41598-021-89710-6] [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: 11/20/2020] [Accepted: 04/27/2021] [Indexed: 11/08/2022] Open
Abstract
Flow conditions have been shown to be important in improving longevity and functionality of primary hepatocytes, but the impact of flow on HepaRG cells is largely unknown. We studied the expression of genes encoding CYP enzymes and transporter proteins and CYP1 and CYP3A4 activity during 8 weeks of culture in HepaRG cells cultured under static conditions (conventional 24-/96-well plate culture with common bicarbonate/CO2 buffering) and under flow conditions in an organ-on-chip (OOC) device. Since the OOC-device is a closed system, bicarbonate/CO2 buffering was not possible, requiring application of another buffering agent, such as HEPES. In order to disentangle the effects of HEPES from the effects of flow, we also applied HEPES-supplemented medium in static cultures and studied gene expression and CYP activity. We found that cells cultured under flow conditions in the OOC-device, as well as cells cultured under static conditions with HEPES-supplemented medium, showed more stable gene expression levels. Furthermore, only cells cultured in the OOC-device showed relatively high baseline CYP1 activity, and their gene expression levels of selected CYPs and transporters were most similar to gene expression levels in human primary hepatocytes. However, there was a decrease in baseline CYP3A4 activity under flow conditions compared to HepaRG cells cultured under static conditions. Altogether, the present study shows that HepaRG cells cultured in the OOC-device were more stable than in static cultures, being a promising in vitro model to study hepatoxicity of chemicals upon chronic exposure.
Collapse
Affiliation(s)
- Loes P M Duivenvoorde
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
| | - Jochem Louisse
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Nicole E T Pinckaers
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Tien Nguyen
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Meike van der Zande
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| |
Collapse
|
24
|
Shibuya K, Watanabe M, Goto R, Zaitsu M, Ganchiku Y, Taketomi A. The Efficacy of the Hepatocyte Spheroids for Hepatocyte Transplantation. Cell Transplant 2021; 30:9636897211000014. [PMID: 33900126 PMCID: PMC8085376 DOI: 10.1177/09636897211000014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The safety and short-term efficacy of hepatocyte transplantation (HCTx) have been widely proven. However, issues such as reduced viability and/or function of hepatocytes, insufficient engraftment, and lack of a long-term effect have to be overcome for widespread application of HCTx. In this study, we evaluated hepatocyte spheroids (HSs), formed by self-aggregation of hepatocytes, as an alternative to hepatocytes in single-cell suspension. Hepatocytes were isolated from C57BL/6 J mice liver using a three-step collagenase perfusion technique and HSs were formed by the hanging drop method. After the spheroids formation, the HSs showed significantly higher mRNA expression of albumin, ornithine transcarbamylase, glucose-6-phosphate, alpha-1-antitrypsin, low density lipoprotein receptor, coagulation factors, and apolipoprotein E (ApoE) than 2 dimensional (2D)-cultured hepatocytes (p < 0.05). Albumin production by HSs was significantly higher than that by 2D-cultured hepatocytes (9.5 ± 2.5 vs 3.5 ± 1.8 μg/dL, p < 0.05). The HSs, but not single hepatocytes, maintained viability and albumin mRNA expression in suspension (92.0 ± 2.8% and 1.03 ± 0.09 at 6 h). HSs (3.6 × 106 cells) or isolated hepatocytes (fSH, 3.6 × 106 cells) were transplanted into the liver of ApoE knockout (KO-/-) mice via the portal vein. Following transplantation, serum ApoE concentration (ng/mL) of HS-transplanted mice (1w: 63.1 ± 56.7, 4w: 17.0 ± 10.9) was higher than that of fSH-transplanted mice (1 w: 33.4 ± 13.0, 4w: 13.7 ± 9.6). In both groups, the mRNA levels of pro-inflammatory cytokines (IL-6, IL-1β, TNF-α, MCP-1, and MIP-1β) were upregulated in the liver following transplantation; however, no significant differences were observed. Pathologically, transplanted HSs were observed as flat cell clusters in contact with the portal vein wall on day 7. Additionally, ApoE positive cells were observed in the liver parenchyma distant from the portal vein on day 28. Our results indicate that HS is a promising alternative to single hepatocytes and can be applied for HCTx.
Collapse
Affiliation(s)
- Kazuaki Shibuya
- Department of Gastroenterological surgery I, 12810Hokkaido university graduate school, kita-ku, Sapporo, Japan
| | - Masaaki Watanabe
- Transplant surgery, 163693Hokkaido University Hospital, kita-ku, Sapporo, Japan
| | - Ryoichi Goto
- Department of Gastroenterological surgery I, 12810Hokkaido university graduate school, kita-ku, Sapporo, Japan
| | - Masaaki Zaitsu
- Department of Gastroenterological surgery I, 12810Hokkaido university graduate school, kita-ku, Sapporo, Japan
| | - Yoshikazu Ganchiku
- Department of Gastroenterological surgery I, 12810Hokkaido university graduate school, kita-ku, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological surgery I, 12810Hokkaido university graduate school, kita-ku, Sapporo, Japan
| |
Collapse
|
25
|
Latest Updates on the Advancement of Polymer-Based Biomicroelectromechanical Systems for Animal Cell Studies. ADVANCES IN POLYMER TECHNOLOGY 2021. [DOI: 10.1155/2021/8816564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Biological sciences have reached the fundamental unit of life: the cell. Ever-growing field of Biological Microelectromechanical Systems (BioMEMSs) is providing new frontiers in both fundamental cell research and various practical applications in cell-related studies. Among various functions of BioMEMS devices, some of the most fundamental processes that can be carried out in such platforms include cell sorting, cell separation, cell isolation or trapping, cell pairing, cell-cell communication, cell differentiation, cell identification, and cell culture. In this article, we review each mentioned application in great details highlighting the latest advancements in fabrication strategy, mechanism of operation, and application of these tools. Moreover, the review article covers the shortcomings of each specific application which can open windows of opportunity for improvement of these devices.
Collapse
|
26
|
Duzagac F, Saorin G, Memeo L, Canzonieri V, Rizzolio F. Microfluidic Organoids-on-a-Chip: Quantum Leap in Cancer Research. Cancers (Basel) 2021; 13:737. [PMID: 33578886 PMCID: PMC7916612 DOI: 10.3390/cancers13040737] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Organ-like cell clusters, so-called organoids, which exhibit self-organized and similar organ functionality as the tissue of origin, have provided a whole new level of bioinspiration for ex vivo systems. Microfluidic organoid or organs-on-a-chip platforms are a new group of micro-engineered promising models that recapitulate 3D tissue structure and physiology and combines several advantages of current in vivo and in vitro models. Microfluidics technology is used in numerous applications since it allows us to control and manipulate fluid flows with a high degree of accuracy. This system is an emerging tool for understanding disease development and progression, especially for personalized therapeutic strategies for cancer treatment, which provide well-grounded, cost-effective, powerful, fast, and reproducible results. In this review, we highlight how the organoid-on-a-chip models have improved the potential of efficiency and reproducibility of organoid cultures. More widely, we discuss current challenges and development on organoid culture systems together with microfluidic approaches and their limitations. Finally, we describe the recent progress and potential utilization in the organs-on-a-chip practice.
Collapse
Affiliation(s)
- Fahriye Duzagac
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Gloria Saorin
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy;
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, 30123 Venezia, Italy; (F.D.); (G.S.)
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
27
|
Yazdian Kashani S, Keshavarz Moraveji M, Taghipoor M, Kowsari-Esfahan R, Hosseini AA, Montazeri L, Dehghan MM, Gholami H, Farzad-Mohajeri S, Mehrjoo M, Majidi M, Renaud P, Bonakdar S. An integrated microfluidic device for stem cell differentiation based on cell-imprinted substrate designed for cartilage regeneration in a rabbit model. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 121:111794. [PMID: 33579444 DOI: 10.1016/j.msec.2020.111794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/30/2020] [Accepted: 12/02/2020] [Indexed: 01/12/2023]
Abstract
Separating cells from the body and cultivating them in vitro will alter the function of cells. Therefore, for optimal cell culture in the laboratory, conditions similar to those of their natural growth should be provided. In previous studies, it has been shown that the use of cellular shape at the culture surface can regulate cellular function. In this work, the efficiency of the imprinting method increased by using microfluidic chip design and fabrication. In this method, first, a cell-imprinted substrate of chondrocytes was made using a microfluidic chip. Afterwards, stem cells were cultured on a cell-imprinted substrate using a second microfluidic chip aligned with the substrate. Therefore, stem cells were precisely placed on the chondrocyte patterns on the substrate and their fibroblast-like morphology was changed to chondrocyte's spherical morphology after 14-days culture in the chip without using any chemical growth factor. After chondrogenic differentiation and in vitro assessments (real-time PCR and immunocytotoxicity), differentiated stem cells were transferred on a collagen-hyaluronic acid scaffold and transplanted in articular cartilage defect of the rabbit. After 6 months, the post-transplantation analysis showed that the articular cartilage defect had been successfully regenerated in differentiated stem cell groups in comparison with the controls. In conclusion, this study showed the potency of the imprinting method for inducing chondrogenicity in stem cells, which can be used in clinical trials due to the safety of the procedure.
Collapse
Affiliation(s)
- Sepideh Yazdian Kashani
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), 1591634311 Tehran, Iran
| | - Mostafa Keshavarz Moraveji
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), 1591634311 Tehran, Iran.
| | - Mojtaba Taghipoor
- School of Mechanical Engineering, Sharif University of Technology, 11155-9567 Tehran, Iran
| | - Reza Kowsari-Esfahan
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran
| | | | - Leila Montazeri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Institute of Biomedical Research, University of Tehran, Tehran, Iran; Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hossein Gholami
- Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Saeed Farzad-Mohajeri
- Institute of Biomedical Research, University of Tehran, Tehran, Iran; Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Morteza Mehrjoo
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran
| | - Mohammad Majidi
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran
| | - Philippe Renaud
- Laboratory of Microsystems (LMIS4), École Polytechnique FÉdÉrale de Lausanne, Station 17, CH-1015 Lausanne, Switzerland
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, P.O. Box 13169-43551, Tehran, Iran.
| |
Collapse
|
28
|
Gupta R, Schrooders Y, Hauser D, van Herwijnen M, Albrecht W, Ter Braak B, Brecklinghaus T, Castell JV, Elenschneider L, Escher S, Guye P, Hengstler JG, Ghallab A, Hansen T, Leist M, Maclennan R, Moritz W, Tolosa L, Tricot T, Verfaillie C, Walker P, van de Water B, Kleinjans J, Caiment F. Comparing in vitro human liver models to in vivo human liver using RNA-Seq. Arch Toxicol 2020; 95:573-589. [PMID: 33106934 PMCID: PMC7870774 DOI: 10.1007/s00204-020-02937-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/12/2020] [Indexed: 01/29/2023]
Abstract
The liver plays an important role in xenobiotic metabolism and represents a primary target for toxic substances. Many different in vitro cell models have been developed in the past decades. In this study, we used RNA-sequencing (RNA-Seq) to analyze the following human in vitro liver cell models in comparison to human liver tissue: cancer-derived cell lines (HepG2, HepaRG 3D), induced pluripotent stem cell-derived hepatocyte-like cells (iPSC-HLCs), cancerous human liver-derived assays (hPCLiS, human precision cut liver slices), non-cancerous human liver-derived assays (PHH, primary human hepatocytes) and 3D liver microtissues. First, using CellNet, we analyzed whether these liver in vitro cell models were indeed classified as liver, based on their baseline expression profile and gene regulatory networks (GRN). More comprehensive analyses using non-differentially expressed genes (non-DEGs) and differential transcript usage (DTU) were applied to assess the coverage for important liver pathways. Through different analyses, we noticed that 3D liver microtissues exhibited a high similarity with in vivo liver, in terms of CellNet (C/T score: 0.98), non-DEGs (10,363) and pathway coverage (highest for 19 out of 20 liver specific pathways shown) at the beginning of the incubation period (0 h) followed by a decrease during long-term incubation for 168 and 336 h. PHH also showed a high degree of similarity with human liver tissue and allowed stable conditions for a short-term cultivation period of 24 h. Using the same metrics, HepG2 cells illustrated the lowest similarity (C/T: 0.51, non-DEGs: 5623, and pathways coverage: least for 7 out of 20) with human liver tissue. The HepG2 are widely used in hepatotoxicity studies, however, due to their lower similarity, they should be used with caution. HepaRG models, iPSC-HLCs, and hPCLiS ranged clearly behind microtissues and PHH but showed higher similarity to human liver tissue than HepG2 cells. In conclusion, this study offers a resource of RNA-Seq data of several biological replicates of human liver cell models in vitro compared to human liver tissue.
Collapse
Affiliation(s)
- Rajinder Gupta
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Yannick Schrooders
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Duncan Hauser
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Wiebke Albrecht
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), Dortmund, Germany
| | - Bas Ter Braak
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9503, 2300 RA, Leiden, The Netherlands
| | - Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), Dortmund, Germany
| | - Jose V Castell
- Instituto de Investigación Sanitaria La Fe, Experimental Hepatology Unit, Valencia, Spain
| | - Leroy Elenschneider
- Fraunhofer Institute for Toxicology and Experimental Medicine Preclinical Pharmacology and In-Vitro Toxicology, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Sylvia Escher
- Fraunhofer Institute for Toxicology and Experimental Medicine Preclinical Pharmacology and In-Vitro Toxicology, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Patrick Guye
- InSphero AG, Wagistrasse 27, 8952, Schlieren, Switzerland
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), Dortmund, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), Dortmund, Germany
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, 83523, Egypt
| | - Tanja Hansen
- Fraunhofer Institute for Toxicology and Experimental Medicine Preclinical Pharmacology and In-Vitro Toxicology, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated, Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Richard Maclennan
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | | | - Laia Tolosa
- Instituto de Investigación Sanitaria La Fe, Unidad Hepatología Experimental, Valencia, Spain
| | - Tine Tricot
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Paul Walker
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9503, 2300 RA, Leiden, The Netherlands
| | - Jos Kleinjans
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
29
|
Moradi E, Jalili-Firoozinezhad S, Solati-Hashjin M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater 2020; 116:67-83. [PMID: 32890749 DOI: 10.1016/j.actbio.2020.08.041] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/08/2023]
Abstract
The liver is the largest internal organ of the body with complex microarchitecture and function that plays critical roles in drug metabolism. Hepatotoxicity and drug-induced liver injury (DILI) caused by various drugs is the main reason for late-stage drug failures. Moreover, liver diseases are among the leading causes of death in the world, with the number of new cases arising each year. Although animal models have been used to understand human drug metabolism and toxicity before clinical trials, tridimensional microphysiological systems, such as liver-on-a-chip (Liver Chip) platforms, could better recapitulate features of human liver physiology and pathophysiology and thus, are often more predictive of human outcome. Liver Chip devices have shown promising results in mimicking in vivo condition by recapitulating the sinusoidal structure of the liver, maintaining high cell viability and cellular phenotypes, and emulating native liver functions. Here, we first review the cellular constituents and physiology of the liver and then critically discuss the state-of-the-art chip-based liver models and their applications in drug screening, disease modeling, and regenerative medicine. We finally address the pending issues of existing platforms and touch upon future directions for developing new, advanced on-chip models.
Collapse
Affiliation(s)
- Ehsanollah Moradi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Mehran Solati-Hashjin
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran.
| |
Collapse
|
30
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
31
|
Chen Z, Zilberberg J, Lee W. Pumpless microfluidic device with open top cell culture under oscillatory shear stress. Biomed Microdevices 2020; 22:58. [PMID: 32833129 DOI: 10.1007/s10544-020-00515-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Here we developed a 96-well plate-based pumpless microfluidic device to mimic bidirectional oscillatory shear stress experienced by osteoblasts at the endosteal niche located at the interface between bone and bone marrow. The culture device was designed to be high-throughput with 32 open top culture chambers for convenient cell seeding and staining. Mathematical modeling was used to simulate the control of oscillatory shear stress with the peak stress in the range of 0.3 to 50 mPa. Osteoblasts, cultured under oscillatory shear stress, were found to be highly viable and significantly aligned along the direction of flow. The modeling and experimental results demonstrate for the first time that cells can be cultured under controllable oscillatory shear stress in the open top culture chamber and pumpless configurations.
Collapse
Affiliation(s)
- Zhehuan Chen
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA
| | - Jenny Zilberberg
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Woo Lee
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA.
| |
Collapse
|
32
|
Ong LJY, Zhu L, Tan GJS, Toh YC. Quantitative Image-Based Cell Viability (QuantICV) Assay for Microfluidic 3D Tissue Culture Applications. MICROMACHINES 2020; 11:mi11070669. [PMID: 32660019 PMCID: PMC7407956 DOI: 10.3390/mi11070669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 01/01/2023]
Abstract
Microfluidic 3D tissue culture systems are attractive for in vitro drug testing applications due to the ability of these platforms to generate 3D tissue models and perform drug testing at a very small scale. However, the minute cell number and liquid volume impose significant technical challenges to perform quantitative cell viability measurements using conventional colorimetric or fluorometric assays, such as MTS or Alamar Blue. Similarly, live-dead staining approaches often utilize metabolic dyes that typically label the cytoplasm of live cells, which makes it difficult to segment and count individual cells in compact 3D tissue cultures. In this paper, we present a quantitative image-based cell viability (QuantICV) assay technique that circumvents current challenges of performing the quantitative cell viability assay in microfluidic 3D tissue cultures. A pair of cell-impermeant nuclear dyes (EthD-1 and DAPI) were used to sequentially label the nuclei of necrotic and total cell populations, respectively. Confocal microscopy and image processing algorithms were employed to visualize and quantify the cell nuclei in the 3D tissue volume. The QuantICV assay was validated and showed good concordance with the conventional bulk MTS assay in static 2D and 3D tumor cell cultures. Finally, the QuantICV assay was employed as an on-chip readout to determine the differential dose responses of parental and metastatic 3D oral squamous cell carcinoma (OSCC) to Gefitinib in a microfluidic 3D culture device. This proposed technique can be useful in microfluidic cell cultures as well as in a situation where conventional cell viability assays are not available.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, #14-01, Singapore 117599, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Level 7, O Block, Gardens Point Campus, Brisbane City QLD 4000, Australia
| | - Liang Zhu
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Singapore Institute of Manufacturing Technology, 31 Biopolis Way, #04-10 Nanos, Singapore 138669, Singapore
- The N.1 Institute for Health, 28 Medical Drive, #05-corridor, Singapore 117456, Singapore
| | - Gabriel Jenn Sern Tan
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, Singapore 117583, Singapore; (L.J.Y.O.); (L.Z.); (G.J.S.T.)
- Institute for Health Innovation and Technology, National University of Singapore, 14 Medical Drive, #14-01, Singapore 117599, Singapore
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Level 7, O Block, Gardens Point Campus, Brisbane City QLD 4000, Australia
- The N.1 Institute for Health, 28 Medical Drive, #05-corridor, Singapore 117456, Singapore
- NUS Tissue Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Q Block-IHBI, 60 Musk Avenue, Kelvin Grove QLD 4059, Australia
- Correspondence:
| |
Collapse
|
33
|
Burton L, Scaife P, Paine SW, Mellor HR, Abernethy L, Littlewood P, Rauch C. Hydrostatic pressure regulates CYP1A2 expression in human hepatocytes via a mechanosensitive aryl hydrocarbon receptor-dependent pathway. Am J Physiol Cell Physiol 2020; 318:C889-C902. [PMID: 32159360 PMCID: PMC7294326 DOI: 10.1152/ajpcell.00472.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Approximately 75% of xenobiotics are primarily eliminated through metabolism; thus the accurate scaling of metabolic clearance is vital to successful drug development. Yet, when data is scaled from in vitro to in vivo, hepatic metabolic clearance, the primary source of metabolism, is still commonly underpredicted. Over the past decades, with biophysics used as a key component to restore aspects of the in vivo environment, several new cell culture settings have been investigated to improve hepatocyte functionalities. Most of these studies have focused on shear stress, i.e., flow mediated by a pressure gradient. One potential conclusion of these studies is that hepatocytes are naturally "mechanosensitive," i.e., they respond to a change in their biophysical environment. We demonstrate that hepatocytes also respond to an increase in hydrostatic pressure that, we suggest, is directly linked to the lobule geometry and vessel density. Furthermore, we demonstrate that hydrostatic pressure improves albumin production and increases cytochrome P-450 (CYP) 1A2 expression levels in an aryl hydrocarbon-dependent manner in human hepatocytes. Increased albumin production and CYP function are commonly attributed to the impacts of shear stress in microfluidic experiments. Therefore, our results highlight evidence of a novel link between hydrostatic pressure and CYP metabolism and demonstrate that the spectrum of hepatocyte mechanosensitivity might be larger than previously thought.
Collapse
Affiliation(s)
- Lewis Burton
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Paula Scaife
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Derby, United Kingdom
| | - Stuart W Paine
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| | - Howard R Mellor
- Vertex Pharmaceuticals Europe Ltd., Abingdon Oxfordshire, United Kingdom
| | - Lynn Abernethy
- Vertex Pharmaceuticals Europe Ltd., Abingdon Oxfordshire, United Kingdom
| | - Peter Littlewood
- Vertex Pharmaceuticals Europe Ltd., Abingdon Oxfordshire, United Kingdom
| | - Cyril Rauch
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, United Kingdom
| |
Collapse
|
34
|
Guzzi F, Candeloro P, Coluccio ML, Cristiani CM, Parrotta EI, Scaramuzzino L, Scalise S, Dattola E, D’Attimo MA, Cuda G, Lamanna E, Passacatini LC, Carbone E, Krühne U, Di Fabrizio E, Perozziello G. A Disposable Passive Microfluidic Device for Cell Culturing. BIOSENSORS-BASEL 2020; 10:bios10030018. [PMID: 32121446 PMCID: PMC7146476 DOI: 10.3390/bios10030018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/21/2020] [Accepted: 02/26/2020] [Indexed: 12/30/2022]
Abstract
In this work, a disposable passive microfluidic device for cell culturing that does not require any additional/external pressure sources is introduced. By regulating the height of fluidic columns and the aperture and closure of the source wells, the device can provide different media and/or drug flows, thereby allowing different flow patterns with respect to time. The device is made of two Polymethylmethacrylate (PMMA) layers fabricated by micro-milling and solvent assisted bonding and allows us to ensure a flow rate of 18.6 μl/ℎ - 7%/day, due to a decrease of the fluid height while the liquid is driven from the reservoirs into the channels. Simulations and experiments were conducted to characterize flows and diffusion in the culture chamber. Melanoma tumor cells were used to test the device and carry out cell culturing experiments for 48 hours. Moreover, HeLa, Jurkat, A549 and HEK293T cell lines were cultivated successfully inside the microfluidic device for 72 hours.
Collapse
Affiliation(s)
- Francesco Guzzi
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Patrizio Candeloro
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Maria Laura Coluccio
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Costanza Maria Cristiani
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Luana Scaramuzzino
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Elisabetta Dattola
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Maria Antonia D’Attimo
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Ernesto Lamanna
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Lucia Carmela Passacatini
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Ennio Carbone
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
| | - Ulrich Krühne
- Department of Chemical and Biochemical Engineering, Technology University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Enzo Di Fabrizio
- Physical Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal 23955-6900, Saudi Arabia;
| | - Gerardo Perozziello
- Department of Experimental and Clinical Medicine, University of Catanzaro, Germaneto, 88100 Catanzaro, Italy; (F.G.); (P.C.); (M.L.C.); (C.M.C.); (E.I.P.); (L.S.); (S.S.); (E.D.); (M.A.D.); (G.C.); (E.L.); (L.C.P.); (E.C.)
- Correspondence:
| |
Collapse
|
35
|
Eluru G, Adhikari JV, Chanda P, Gorthi SS. Hand-Powered Elastomeric Pump for Microfluidic Point-of-Care Diagnostics. MICROMACHINES 2020; 11:mi11010067. [PMID: 31936146 PMCID: PMC7019644 DOI: 10.3390/mi11010067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 01/05/2023]
Abstract
The pumping of fluids into microfluidic channels has become almost an unavoidable operation in all microfluidic applications. Such a need has seen an outburst of several techniques for pumping, out of which the majority of techniques involve complicated fabrication, as they require the introduction of electrodes, valves, piezoelectric materials, acoustic transducers, etc., into the microfluidic device. In addition to the complexity, this also escalates the cost incurred per device. Further, the use of stable external power supplies to produce such a pumping action adds to the bulkiness of the pumps, making them unsuitable for point-of-care diagnostic (POCD) applications. This paper reports a technique of pumping that is simple to realize and does not require external electric/magnetic power, but exploits the elastic properties of materials to achieve the pumping action. This mechanism of pumping ensured the cost per pump to less than 4 USD and can be used for at least 500 times. Several simulations, validation, and characterization experiments were performed on the developed pump to establish its functionality and suitability for use in POCD applications.
Collapse
|
36
|
Fattahi P, Haque A, Son KJ, Guild J, Revzin A. Microfluidic devices, accumulation of endogenous signals and stem cell fate selection. Differentiation 2019; 112:39-46. [PMID: 31884176 DOI: 10.1016/j.diff.2019.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/06/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Pouria Fattahi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Amranul Haque
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kyung Jin Son
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Joshua Guild
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
37
|
Abstract
Engineering approaches were adopted for liver microsystems to recapitulate cell arrangements and culture microenvironments in vivo for sensitive, high-throughput and biomimetic drug screening. This review introduces liver microsystems in vitro for drug hepatotoxicity, drug-drug interactions, metabolic function and enzyme induction, based on cell micropatterning, hydrogel biofabrication and microfluidic perfusion. The engineered microsystems provide varied microenvironments for cell culture that feature cell coculture with non-parenchymal cells, in a heterogeneous extracellular matrix and under controllable perfusion. The engineering methods described include cell micropatterning with soft lithography and dielectrophoresis, hydrogel biofabrication with photolithography, micromolding and 3D bioprinting, and microfluidic perfusion with endothelial-like structures and gradient generators. We discuss the major challenges and trends of liver microsystems to study drug response in vitro.
Collapse
Affiliation(s)
- Jyong-Huei Lee
- Department of Mechanical Engineering, National Taiwan University, Taipei, Taiwan
| | - Kuan-Lun Ho
- Department of Mechanical Engineering, National Taiwan University, Taipei, Taiwan
| | - Shih-Kang Fan
- Department of Mechanical and Nuclear Engineering, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
38
|
Ong LJY, Ching T, Chong LH, Arora S, Li H, Hashimoto M, DasGupta R, Yuen PK, Toh YC. Self-aligning Tetris-Like (TILE) modular microfluidic platform for mimicking multi-organ interactions. LAB ON A CHIP 2019; 19:2178-2191. [PMID: 31179467 DOI: 10.1039/c9lc00160c] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Multi-organ perfusion systems offer the unique opportunity to mimic different physiological systemic interactions. However, existing multi-organ culture platforms have limited flexibility in specifying the culture conditions, device architectures, and fluidic connectivity simultaneously. Here, we report a modular microfluidic platform that addresses this limitation by enabling easy conversion of existing microfluidic devices into tissue and fluid control modules with self-aligning magnetic interconnects. This enables a 'stick-n-play' approach to assemble planar perfusion circuits that are amenable to both bioimaging-based and analytical measurements. A myriad of tissue culture and flow control TILE modules were successfully constructed with backward compatibility. Finally, we demonstrate applications in constructing recirculating multi-organ systems to emulate liver-mediated bioactivation of nutraceuticals and prodrugs to modulate their therapeutic efficacies in the context of atherosclerosis and cancer. This platform greatly facilitates the integration of existing organs-on-chip models to provide an intuitive and flexible way for users to configure different multi-organ perfusion systems.
Collapse
Affiliation(s)
- Louis Jun Ye Ong
- Department of Biomedical Engineering, National University of Singapore, 4, Engineering Drive 3, E4-04-10, 117583, Singapore.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sun M, Wong JY, Nugraha B, Ananthanarayanan A, Liu Z, Lee F, Gupta K, Fong EL, Huang X, Yu H. Cleavable cellulosic sponge for functional hepatic cell culture and retrieval. Biomaterials 2019; 201:16-32. [DOI: 10.1016/j.biomaterials.2019.01.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/18/2019] [Accepted: 01/20/2019] [Indexed: 12/27/2022]
|
40
|
Natale A, Vanmol K, Arslan A, Van Vlierberghe S, Dubruel P, Van Erps J, Thienpont H, Buzgo M, Boeckmans J, De Kock J, Vanhaecke T, Rogiers V, Rodrigues RM. Technological advancements for the development of stem cell-based models for hepatotoxicity testing. Arch Toxicol 2019; 93:1789-1805. [PMID: 31037322 DOI: 10.1007/s00204-019-02465-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Stem cells are characterized by their self-renewal capacity and their ability to differentiate into multiple cell types of the human body. Using directed differentiation strategies, stem cells can now be converted into hepatocyte-like cells (HLCs) and therefore, represent a unique cell source for toxicological applications in vitro. However, the acquired hepatic functionality of stem cell-derived HLCs is still significantly inferior to primary human hepatocytes. One of the main reasons for this is that most in vitro models use traditional two-dimensional (2D) setups where the flat substrata cannot properly mimic the physiology of the human liver. Therefore, 2D-setups are progressively being replaced by more advanced culture systems, which attempt to replicate the natural liver microenvironment, in which stem cells can better differentiate towards HLCs. This review highlights the most recent cell culture systems, including scaffold-free and scaffold-based three-dimensional (3D) technologies and microfluidics that can be employed for culture and hepatic differentiation of stem cells intended for hepatotoxicity testing. These methodologies have shown to improve in vitro liver cell functionality according to the in vivo liver physiology and allow to establish stem cell-based hepatic in vitro platforms for the accurate evaluation of xenobiotics.
Collapse
Affiliation(s)
- Alessandra Natale
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Koen Vanmol
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Aysu Arslan
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group (PBM), Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Jürgen Van Erps
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | - Hugo Thienpont
- Brussels Photonics (B-PHOT), Vrije Universiteit Brussel and Flanders Make, Brussels, Belgium
| | | | - Joost Boeckmans
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Joery De Kock
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Tamara Vanhaecke
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vera Rogiers
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium
| | - Robim M Rodrigues
- Department of In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
41
|
Ahn J, Lee HJ, Oh SJ, Kim W, Mun SJ, Lee JH, Jung CR, Cho HS, Kim DS, Son MJ, Chung KS. Developing scalable cultivation systems of hepatic spheroids for drug metabolism via genomic and functional analyses. Biotechnol Bioeng 2019; 116:1496-1508. [PMID: 30737956 DOI: 10.1002/bit.26954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/18/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Spheroids, a widely used three-dimensional (3D) culture model, are standard in hepatocyte culture as they preserve long-term hepatocyte functionality and enhance survivability. In this study, we investigated the effects of three operation modes in 3D culture - static, orbital shaking, and under vertical bidirectional flow using spheroid forming units (SFUs) on hepatic differentiation and drug metabolism to propose the best for mass production of functionally enhanced spheroids. Spheroids in SFUs exhibited increased hepatic gene expression, albumin secretion, and cytochrome P450 3A4 (CYP3A4) activity during the differentiation period (12 days). SFUs advantages include facilitated mass production and a relatively earlier peak of CYP3A4 activity. However, CYP3A4 activity was not well maintained under dimethyl sulfoxide (DMSO)-free conditions (13-18 days), dramatically reducing drug metabolism capability. Continued shear stimulation without differentiation stimuli in assay conditions markedly attenuated CYP3A4 activity, which was less severe in static conditions. In this condition, SFU spheroids exhibited dedifferentiation characteristics, such as increased proliferation and Notch signaling genes. We found that the dedifferentiation could be overcome by using the serum-free medium formulation. Therefore, we suggest that SFUs represent the best option for the mass production of functionally improved spheroids and so the serum-free conditions should be maintained during drug metabolism analysis.
Collapse
Affiliation(s)
- Jiwon Ahn
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ho-Joon Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Soo Jin Oh
- New Drug Development Center, Asan Medical Center and Convergence Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Wantae Kim
- Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea
| | - Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jae-Hye Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rock Jung
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Gene Therapy Unit, KRIBB, Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Dae-Soo Kim
- Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Genome Research Center, KRIBB, Daejeon, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyung-Sook Chung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Biomedical Translational Research Center, KRIBB, Daejeon, Republic of Korea.,Functional Genomics, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
42
|
Liu R, Yao X, Liu X, Ding J. Proliferation of Cells with Severe Nuclear Deformation on a Micropillar Array. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:284-299. [PMID: 30513205 DOI: 10.1021/acs.langmuir.8b03452] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cellular responses on a topographic surface are fundamental topics about interfaces and biology. Herein, a poly(lactide- co-glycolide) (PLGA) micropillar array was prepared and found to trigger significant self-deformation of cell nuclei. The time-dependent cell viability and thus cell proliferation was investigated. Despite significant nuclear deformation, all of the examined cell types (Hela, HepG2, MC3T3-E1, and NIH3T3) could survive and proliferate on the micropillar array yet exhibited different proliferation abilities. Compared to the corresponding groups on the smooth surface, the cell proliferation abilities on the micropillar array were decreased for Hela and MC3T3-E1 cells and did not change significantly for HepG2 and NIH3T3 cells. We also found that whether the proliferation ability changed was related to whether the nuclear sizes decreased in the micropillar array, and thus the size deformation of cell nuclei should, besides shape deformation, be taken into consideration in studies of cells on topological surfaces.
Collapse
Affiliation(s)
- Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiang Yao
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiangnan Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| |
Collapse
|
43
|
Chong LH, Li H, Wetzel I, Cho H, Toh YC. A liver-immune coculture array for predicting systemic drug-induced skin sensitization. LAB ON A CHIP 2018; 18:3239-3250. [PMID: 30252012 DOI: 10.1039/c8lc00790j] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Drug-induced skin sensitization is prevalent worldwide and can trigger life-threatening health conditions, such as Stevens Johnson Syndrome. However, existing in vitro skin models cannot adequately predict the skin sensitization effects of drugs administered into the systemic circulation because dermal inflammation and injury are preceded by conversion of parent drugs into antigenic reactive metabolites in the liver and subsequent activation of the immune system. Here, we demonstrate that recapitulation of these early tandem cellular processes in a compartmentalized liver-immune coculture array is sufficient to predict the skin sensitization potential of systemic drugs. Human progenitor cell (HepaRG)-derived hepatocyte spheroids and U937 myeloid cells, a representative antigen presenting cell (APC), can maintain their respective functions in 2 concentric micro-chambers, which are connected by a diffusion microchannel network. Paradigm drugs that are reported to cause severe cutaneous drug reactions (i.e. carbamazepine, phenytoin and allopurinol) can be metabolized into their reactive metabolites, which diffuse efficiently into the adjoining immune compartment within a 48 hour period. By measuring the extent of U937 activation as indicated by IL8, IL1β and CD86 upregulation upon drug administration, we show that the liver-immune coculture array more consistently and reliably distinguish all 3-paradigm skin sensitizing drugs from a non-skin sensitizer than conventional bulk Transwell coculture. Given its miniaturized format, design simplicity and prediction capability, this novel in vitro system can be readily scaled into a screenable platform to identify the skin sensitization potential of systemically-administered drugs.
Collapse
Affiliation(s)
- Lor Huai Chong
- Department of Biomedical Engineering, National University of Singapore, Singapore.
| | | | | | | | | |
Collapse
|
44
|
Lim J, Tahk D, Yu J, Min DH, Jeon NL. Design rules for a tunable merged-tip microneedle. MICROSYSTEMS & NANOENGINEERING 2018; 4:29. [PMID: 31057917 PMCID: PMC6220166 DOI: 10.1038/s41378-018-0028-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/04/2018] [Accepted: 06/24/2018] [Indexed: 05/05/2023]
Abstract
This publication proposes the use of an elasto-capillarity-driven self-assembly for fabricating a microscale merged-tip structure out of a variety of biocompatible UV-curable polymers for use as a microneedle platform. In addition, the novel merged-tip microstructure constitutes a new class of microneedles, which incorporates the convergence of biocompatible polymer micropillars, leading to the formation of a sharp tip and an open cavity capable of both liquid trapping and volume control. When combined with biocompatible photopolymer micropillar arrays fabricated with photolithography, elasto-capillarity-driven self-assembly provides a means for producing a complex microneedle-like structure without the use of micromolding or micromachining. This publication also explores and defines the design rules by which several fabrication aspects, such as micropillar dimensions, shapes, pattern array configurations, and materials, can be manipulated to produce a customizable microneedle array with controllable cavity volumes, fracture points, and merge profiles. In addition, the incorporation of a modular through-hole micropore membrane base was also investigated as a method for constitutive payload delivery and fluid-sampling functionalities. The flexibility and fabrication simplicity of the merged-tip microneedle platform holds promise in transdermal drug delivery applications.
Collapse
Affiliation(s)
- Jungeun Lim
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826 South Korea
- Division of WCU Multiscale Mechanical Design, Seoul National University, Seoul, 08826 South Korea
| | - Dongha Tahk
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826 South Korea
- Division of WCU Multiscale Mechanical Design, Seoul National University, Seoul, 08826 South Korea
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, 08826 South Korea
| | - James Yu
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826 South Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826 South Korea
- Division of WCU Multiscale Mechanical Design, Seoul National University, Seoul, 08826 South Korea
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, 08826 South Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826 South Korea
| |
Collapse
|
45
|
Gill US, Pallett LJ, Kennedy PTF, Maini MK. Liver sampling: a vital window into HBV pathogenesis on the path to functional cure. Gut 2018; 67:767-775. [PMID: 29331944 PMCID: PMC6058064 DOI: 10.1136/gutjnl-2017-314873] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 12/12/2022]
Abstract
In order to optimally refine the multiple emerging drug targets for hepatitis B virus (HBV), it is vital to evaluate virological and immunological changes at the site of infection. Traditionally liver biopsy has been the mainstay of HBV disease assessment, but with the emergence of non-invasive markers of liver fibrosis, there has been a move away from tissue sampling. Here we argue that liver biopsy remains an important tool, not only for the clinical assessment of HBV but also for research progress and evaluation of novel agents. The importance of liver sampling has been underscored by recent findings of specialised subsets of tissue-resident immune subsets capable of efficient pathogen surveillance, compartmentalised in the liver and not sampled in the blood. Importantly, the assessment of virological parameters, such as cccDNA quantitation, also requires access to liver tissue. We discuss strategies to maximise information obtained from the site of infection and disease pathology. Fine needle aspirates of the liver may allow longitudinal sampling of the local virus/host landscape. The careful utilisation of liver tissue and aspirates in conjunction with blood will provide critical information in the assessment of new therapeutics for the functional cure of HBV.
Collapse
Affiliation(s)
- Upkar S Gill
- Department of Hepatology, Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Patrick T F Kennedy
- Department of Hepatology, Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mala K Maini
- Division of Infection and Immunity, UCL, London, UK
| |
Collapse
|
46
|
Ortega-Prieto AM, Skelton JK, Wai SN, Large E, Lussignol M, Vizcay-Barrena G, Hughes D, Fleck RA, Thursz M, Catanese MT, Dorner M. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat Commun 2018; 9:682. [PMID: 29445209 PMCID: PMC5813240 DOI: 10.1038/s41467-018-02969-8] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 01/09/2018] [Indexed: 12/21/2022] Open
Abstract
With more than 240 million people infected, hepatitis B virus (HBV) is a major health concern. The inability to mimic the complexity of the liver using cell lines and regular primary human hepatocyte (PHH) cultures pose significant limitations for studying host/pathogen interactions. Here, we describe a 3D microfluidic PHH system permissive to HBV infection, which can be maintained for at least 40 days. This system enables the recapitulation of all steps of the HBV life cycle, including the replication of patient-derived HBV and the maintenance of HBV cccDNA. We show that innate immune and cytokine responses following infection with HBV mimic those observed in HBV-infected patients, thus allowing the dissection of pathways important for immune evasion and validation of biomarkers. Additionally, we demonstrate that the co-culture of PHH with other non-parenchymal cells enables the identification of the cellular origin of immune effectors, thus providing a valuable preclinical platform for HBV research.
Collapse
Affiliation(s)
- A M Ortega-Prieto
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
| | - J K Skelton
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
| | - S N Wai
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK
- Section of Hepatology, Department of Medicine, Imperial College London, London, W2 1NY, UK
| | - E Large
- CN Bio Innovations Ltd, Welwyn Garden City, AL7 3AX, UK
| | - M Lussignol
- Department of Infectious Diseases, King's College London, London, WC2R 2LS, UK
| | - G Vizcay-Barrena
- Centre For Ultrastructural Imaging, King's College London, London, WC2R 2LS, UK
| | - D Hughes
- CN Bio Innovations Ltd, Welwyn Garden City, AL7 3AX, UK
| | - R A Fleck
- Centre For Ultrastructural Imaging, King's College London, London, WC2R 2LS, UK
| | - M Thursz
- Section of Hepatology, Department of Medicine, Imperial College London, London, W2 1NY, UK
| | - M T Catanese
- Department of Infectious Diseases, King's College London, London, WC2R 2LS, UK
| | - M Dorner
- Section of Virology, Department of Medicine, Imperial College London, London, W2 1PG, UK.
| |
Collapse
|
47
|
Sakai Y, Shinohara M. [Tissue engineering-based approaches to enhance physiological relevancy of cell-based assays]. Nihon Yakurigaku Zasshi 2018; 151:56-61. [PMID: 29415926 DOI: 10.1254/fpj.151.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Animal-free and mechanism-based understanding of human body responses is the ultimate goal of alternative to animal experiments. To achieve this goal, integration of advanced cell-based assays using iPS/ES cell technologies with various numerical methods are required. In this review, from the standpoint of tissue engineering, we focused first on the enhancement of physiological relevance of tissue culture models by overcoming the problem between 3D cellular organization and oxygen/nutrient supply. Second, we summarized the concept and actual systems of microfluidic-based body/organ on-a-chip systems, also called as microphysiological system, MPS, particularly for liver on-a-chip systems. Finally, remaining issues were discussed to realize better physiological relevance in vitro.
Collapse
Affiliation(s)
- Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo.,International Research Center on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo
| | - Marie Shinohara
- International Research Center on Integrative Biomedical Systems (CIBiS), Institute of Industrial Science, The University of Tokyo
| |
Collapse
|
48
|
Pumpless microfluidic system driven by hydrostatic pressure induces and maintains mouse spermatogenesis in vitro. Sci Rep 2017; 7:15459. [PMID: 29133858 PMCID: PMC5684205 DOI: 10.1038/s41598-017-15799-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 11/01/2017] [Indexed: 11/12/2022] Open
Abstract
Three-dimensional aggregation and organ culture methods are critical for recreating in vivo cellular phenomena outside the body. Previously, we used the conventional gas liquid interphase organ culture method to induce complete mouse spermatogenesis. After incorporating microfluidic systems, we achieved a significant increase in efficiency and duration of spermatogenesis. One of the major drawbacks preventing the popularization of microfluidics, however, is the use of a power-pump to generate medium flow. In this study, we produced a pumpless microfluidic device using hydrostatic pressure and a resistance circuit to facilitate slow, longer lasting medium flow. During three months of culture, results in induction and maintenance of spermatogenesis showed no difference between pumpless and pump-driven devices. Correspondingly, the spermatogonial population was favorably maintained in the pumpless device compared to the conventional method. These results show the advantage of using microfluidic systems for organ culture experiments. Our pumpless device could be applied to a variety of other tissues and organs, and may revolutionize organ culture methods as a whole.
Collapse
|
49
|
Ong LJY, Islam A, DasGupta R, Iyer NG, Leo HL, Toh YC. A 3D printed microfluidic perfusion device for multicellular spheroid cultures. Biofabrication 2017; 9:045005. [DOI: 10.1088/1758-5090/aa8858] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|