1
|
Ehlers H, Olivier T, Trietsch SJ, Vulto P, Burton TP, van den Broek LJ. Microfluidic artery-on-a-chip model with unidirectional gravity-driven flow for high-throughput applications. LAB ON A CHIP 2025. [PMID: 40261030 DOI: 10.1039/d4lc01109k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, with a noticeable decline in the approval of new therapeutic interventions. Currently, there is no gold standard for developing new therapies for CVDs, and preclinical models do not translate to clinical efficacy. Therefore, there is an urgent need for in vitro models that more accurately mimic human disease processes. Here we describe a model of the artery consisting of monocultures of human coronary artery endothelial cells (HCAECs) or cocultures of HCAECs with human coronary artery smooth muscle cells (HCASMCs). The model was established in the OrganoPlate® 2-lane-48 UF, a novel microfluidic device, comprised of a microtiter plate footprint with 48 chips. Fluid is circulated in a unidirectional manner by interval rocking. The creation of an air-liquid interface at the inlets at a given inclination is used to select flow paths and establish flow in one direction only, whilst capillary forces ensure the channel remains filled with fluid. We investigated the impact of unidirectional or bidirectional flow conditions. Under unidirectional flow, endothelial cells aligned with the flow direction, decreased fibronectin deposition, and smooth muscle cells presented a non-contractile phenotype, emulating the characteristics of healthy arteries. Contrarily, bidirectional flow mimicked features of early endothelial dysfunction, such as contractile morphology of vessels and increased fibronectin secretion, ICAM-1 staining, and lipid deposits. Vascular inflammation could be induced by the addition of TNFα and IL-1β in both flow conditions. Overall, the OrganoPlate® 2-lane-48 UF is a powerful platform providing both throughput and improved flow control, for creating more physiological models. Its ability to replicate key features of a healthy and diseased artery, its potential use in drug screening, and its compatibility with lab automation make it an invaluable tool for researchers aiming for more accurate and efficient therapeutic development in CVD.
Collapse
Affiliation(s)
- H Ehlers
- Mimetas B.V., Oegstgeest, The Netherlands.
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - T Olivier
- Mimetas B.V., Oegstgeest, The Netherlands.
| | | | - P Vulto
- Mimetas B.V., Oegstgeest, The Netherlands.
| | - T P Burton
- Mimetas B.V., Oegstgeest, The Netherlands.
| | | |
Collapse
|
2
|
Limjanthong N, Sugiura S, Oda T, Takusari F, Fujiwara Y, Miyazaki T, Naganuma K, Ohnuma K. Monitoring and optimization of the microenvironment in a gravity-driven microfluidic system placed on a slow-tilting table. J Biosci Bioeng 2025; 139:311-318. [PMID: 39843315 DOI: 10.1016/j.jbiosc.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Gravity-driven microfluidic chips offer portability and flexibility in different settings because pumps and connecting tubes are unnecessary for driving fluid flow. In a previous study, human induced pluripotent stem cells were cultured using gravity-driven microfluidics, with the liquid flow rate regulated by a tilting table. However, instability in cell culture has been observed, occasionally leading to cell death owing to unknown causes. This study measured the ability of a gravity-driven microfluidic system to maintain essential microenvironments, specifically the flow rate, CO2 levels, temperature, and humidity. The incubation procedure was improved to stabilize the parameters at target values. Improvements in the incubation process reduced the time required to reach the stabilized value for CO2, temperature, and humidity by 85, 67, and 5 %, respectively, compared to previous methods. The system demonstrated a precise flow rate, confirmed by a consistent increase in the downstream tank's medium volume after 4 h of perfusion. In addition, the adjustment of the tilting table maintained a steady angle and effectively regulated the flow rate, with the measured flow rate consistent with the theoretical value. The gravity-driven microfluidic system effectively facilitated the culture and differentiation of human iPSCs into the mesodermal lineage after bone morphogenetic protein 4 induction, as indicated by positive SSEA1 immunostaining, demonstrating its potential for stem cell research. Gravity-driven microfluidic systems satisfy these requirements and are suitable for stem cell culture experiments.
Collapse
Affiliation(s)
- Nuttakrit Limjanthong
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| | - Shinji Sugiura
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, 1-1-1 Higashi, Tsukuba 305-8565, Japan.
| | - Taira Oda
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| | - Fuko Takusari
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| | - Yasunori Fujiwara
- Division of Mechanical and Intelligent Systems Engineering, National Institute of Technology, Ichinoseki College, Aza-Takanashi, Hagisho, Ichinoseki, Iwate 021-8511, Japan.
| | - Toshimasa Miyazaki
- Department of Electrical Electronics and Information Engineering, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| | - Kosei Naganuma
- Environment and Process Design Laboratory, Extreme Energy-Density Research Institute, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| | - Kiyoshi Ohnuma
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan; Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kamitomioka-machi, Nagaoka, Niigata 940-2188, Japan.
| |
Collapse
|
3
|
Agarwal SS, Holter JC, Jones TH, Fuller BT, Tinapple JW, Barlage JM, Song JW. A Modular, Cost-Effective, and Pumpless Perfusion Assembly for the Long-Term Culture of Engineered Microvessels. MICROMACHINES 2025; 16:351. [PMID: 40141962 PMCID: PMC11945127 DOI: 10.3390/mi16030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025]
Abstract
Continuous perfusion is necessary to sustain microphysiological systems and other microfluidic cell cultures. However, most of the established microfluidic perfusion systems, such as syringe pumps, peristaltic pumps, and rocker plates, have several operational challenges and may be cost-prohibitive, especially for laboratories with no microsystems engineering expertise. Here, we address the need for a cost-efficient, easy-to-implement, and reliable microfluidic perfusion system. Our solution is a modular pumpless perfusion assembly (PPA), which is constructed from commercially available, interchangeable, and aseptically packaged syringes and syringe filters. The total cost for the components of each assembled PPA is USD 1-2. The PPA retains the simplicity of gravity-based pumpless flow systems but incorporates high resistance filters that enable slow and sustained flow for extended periods of time (hours to days). The perfusion characteristics of the PPA were determined by theoretical calculations of the total hydraulic resistance of the assembly and experimental characterization of specific filter resistances. We demonstrated that the PPA enabled reliable long-term culture of engineered endothelialized 3-D microvessels for several weeks. Taken together, our novel PPA solution is simply constructed from extremely low-cost and commercially available laboratory supplies and facilitates robust cell culture and compatibility with current microfluidic setups.
Collapse
Affiliation(s)
- Shashwat S. Agarwal
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
| | - Jacob C. Holter
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Travis H. Jones
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
| | - Brendan T. Fuller
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Joseph W. Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA; (J.C.H.); (B.T.F.); (J.W.T.)
| | - Joseph M. Barlage
- Department of Biomedical Education and Anatomy, The Ohio State University, Columbus, OH 43210, USA;
| | - Jonathan W. Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH 43210, USA; (S.S.A.); (T.H.J.)
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Ansarizadeh M, Nguyen HT, Lazovic B, Kettunen J, De Silva L, Sivakumar R, Junttila P, Rissanen SL, Hicks R, Singh P, Eklund L. Microfluidic vessel-on-chip platform for investigation of cellular defects in venous malformations and responses to various shear stress and flow conditions. LAB ON A CHIP 2025; 25:613-630. [PMID: 39847008 DOI: 10.1039/d4lc00824c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
A novel microfluidic platform was designed to study the cellular architecture of endothelial cells (ECs) in an environment replicating the 3D organization and flow of blood vessels. In particular, the platform was constructed to investigate EC defects in slow-flow venous malformations (VMs) under varying shear stress and flow conditions. The platform featured a standard microtiter plate footprint containing 32 microfluidic units capable of replicating wall shear stress (WSS) in normal veins and enabling precise control of shear stress and flow directionality without the need for complex pumping systems. Using genetically engineered human umbilical vein endothelial cells (HUVECs) and induced pluripotent stem cell (iPSC)-derived ECs (iECs) to express the recurrent TIE2L914F VM mutation we assessed responses on EC orientation and area, actin organization, and Golgi polarization to uni- and bidirectional flow and varying WSS. Comparison of control and TIE2L914F expressing ECs showed differential cellular responses to flow and WSS in terms of cell shape elongation, orientation of F-actin, and Golgi polarization, indicating altered mechanosensory or mechanotransduction signaling pathways in the presence of the VM causative mutation. The data also revealed significant differences in how the primary and iPSC-derived iECs responded to flow. As a conclusion, the developed microfluidic platform allowed simulation of multiple flow conditions in a scalable and pumpless format. The design made it a desirable tool for studying different EC types as well as cellular changes in vascular disease. The platform should offer new opportunities for biomechanical research by providing a controlled environment to analyze the flow-dependent mechanosensory pathways in ECs.
Collapse
Affiliation(s)
- Mohammadhassan Ansarizadeh
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| | - Hoang-Tuan Nguyen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
- Finnadvance Ltd., Oulu, Finland
| | - Bojana Lazovic
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Laknee De Silva
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| | | | | | | | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | | | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland.
| |
Collapse
|
5
|
Kim R, Sung JH. Recent Advances in Gut- and Gut-Organ-Axis-on-a-Chip Models. Adv Healthc Mater 2024; 13:e2302777. [PMID: 38243887 DOI: 10.1002/adhm.202302777] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/21/2023] [Indexed: 01/22/2024]
Abstract
The human gut extracts nutrients from the diet while forming the largest barrier against the outer environment. In addition, the gut actively maintains homeostasis through intricate interactions with the gut microbes, the immune system, the enteric nervous system, and other organs. These interactions influence digestive health and, furthermore, play crucial roles in systemic health and disease. Given its primary role in absorbing and metabolizing orally administered drugs, there is significant interest in the development of preclinical in vitro model systems that can accurately emulate the intestine in vivo. A gut-on-a-chip system holds great potential as a testing and screening platform because of its ability to emulate the physiological aspects of in vivo tissues and expandability to incorporate and combine with other organs. This review aims to identify the key physiological features of the human gut that need to be incorporated to build more accurate preclinical models and highlights the recent progress in gut-on-a-chip systems and competing technologies toward building more physiologically relevant preclinical model systems. Furthermore, various efforts to construct multi-organ systems with the gut, called gut-organ-axis-on-a-chip models, are discussed. In vitro gut models with physiological relevance can provide valuable platforms for bridging the gap between preclinical and clinical studies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, 04066, Republic of Korea
| |
Collapse
|
6
|
Zhang F, Lin DSY, Rajasekar S, Sotra A, Zhang B. Pump-Less Platform Enables Long-Term Recirculating Perfusion of 3D Printed Tubular Tissues. Adv Healthc Mater 2023; 12:e2300423. [PMID: 37543836 PMCID: PMC11469154 DOI: 10.1002/adhm.202300423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/13/2023] [Indexed: 08/07/2023]
Abstract
The direction and pattern of fluid flow affect vascular structure and function, in which vessel-lining endothelial cells exhibit variable cellular morphologies and vessel remodeling by mechanosensing. To recapitulate this microenvironment, some approaches have been reported to successfully apply unidirectional flow on endothelial cells in organ-on-a-chip systems. However, these platforms encounter drawbacks such as the dependency on pumps or confinement to closed microfluidic channels. These constraints impede their synergy with advanced biofabrication techniques like 3D bioprinting, thereby curtailing the potential to introduce greater complexity into engineered tissues. Herein, a pumpless recirculating platform (UniPlate) that enables unidirectional media recirculation through 3D printed tubular tissues, is demonstrated.The device is made of polystyrene via injection molding in combination with 3D printed sacrifical gelatin templates. Tubular blood vessels with unidirectional perfusion are firstly engineered. Then the design is expanded to incorporate duo-recirculating flow for culturing vascularized renal proximal tubules with glucose reabsorption function. In addition to media recirculation, human monocyte recirculation in engineered blood vessels is also demonstrated for over 24 h, with minimal loss of cells, cell viability, and inflammatory activation. UniPlate can be a valuable tool to more precisely control the cellular microenvironment of organ-on-a-chip systems for drug discovery.
Collapse
Affiliation(s)
- Feng Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Dawn S. Y. Lin
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | | | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
7
|
Juste-Lanas Y, Hervas-Raluy S, García-Aznar JM, González-Loyola A. Fluid flow to mimic organ function in 3D in vitro models. APL Bioeng 2023; 7:031501. [PMID: 37547671 PMCID: PMC10404142 DOI: 10.1063/5.0146000] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 08/08/2023] Open
Abstract
Many different strategies can be found in the literature to model organ physiology, tissue functionality, and disease in vitro; however, most of these models lack the physiological fluid dynamics present in vivo. Here, we highlight the importance of fluid flow for tissue homeostasis, specifically in vessels, other lumen structures, and interstitium, to point out the need of perfusion in current 3D in vitro models. Importantly, the advantages and limitations of the different current experimental fluid-flow setups are discussed. Finally, we shed light on current challenges and future focus of fluid flow models applied to the newest bioengineering state-of-the-art platforms, such as organoids and organ-on-a-chip, as the most sophisticated and physiological preclinical platforms.
Collapse
Affiliation(s)
| | - Silvia Hervas-Raluy
- Department of Mechanical Engineering, Engineering Research Institute of Aragón (I3A), University of Zaragoza, Zaragoza, Spain
| | | | | |
Collapse
|
8
|
Van Os L, Engelhardt B, Guenat OT. Integration of immune cells in organs-on-chips: a tutorial. Front Bioeng Biotechnol 2023; 11:1191104. [PMID: 37324438 PMCID: PMC10267470 DOI: 10.3389/fbioe.2023.1191104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Viral and bacterial infections continue to pose significant challenges for numerous individuals globally. To develop novel therapies to combat infections, more insight into the actions of the human innate and adaptive immune system during infection is necessary. Human in vitro models, such as organs-on-chip (OOC) models, have proven to be a valuable addition to the tissue modeling toolbox. The incorporation of an immune component is needed to bring OOC models to the next level and enable them to mimic complex biological responses. The immune system affects many (patho)physiological processes in the human body, such as those taking place during an infection. This tutorial review introduces the reader to the building blocks of an OOC model of acute infection to investigate recruitment of circulating immune cells into the infected tissue. The multi-step extravasation cascade in vivo is described, followed by an in-depth guide on how to model this process on a chip. Next to chip design, creation of a chemotactic gradient and incorporation of endothelial, epithelial, and immune cells, the review focuses on the hydrogel extracellular matrix (ECM) to accurately model the interstitial space through which extravasated immune cells migrate towards the site of infection. Overall, this tutorial review is a practical guide for developing an OOC model of immune cell migration from the blood into the interstitial space during infection.
Collapse
Affiliation(s)
- Lisette Van Os
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Olivier T. Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
9
|
Dufva M. A quantitative meta-analysis comparing cell models in perfused organ on a chip with static cell cultures. Sci Rep 2023; 13:8233. [PMID: 37217582 DOI: 10.1038/s41598-023-35043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
As many consider organ on a chip for better in vitro models, it is timely to extract quantitative data from the literature to compare responses of cells under flow in chips to corresponding static incubations. Of 2828 screened articles, 464 articles described flow for cell culture and 146 contained correct controls and quantified data. Analysis of 1718 ratios between biomarkers measured in cells under flow and static cultures showed that the in all cell types, many biomarkers were unregulated by flow and only some specific biomarkers responded strongly to flow. Biomarkers in cells from the blood vessels walls, the intestine, tumours, pancreatic island, and the liver reacted most strongly to flow. Only 26 biomarkers were analysed in at least two different articles for a given cell type. Of these, the CYP3A4 activity in CaCo2 cells and PXR mRNA levels in hepatocytes were induced more than two-fold by flow. Furthermore, the reproducibility between articles was low as 52 of 95 articles did not show the same response to flow for a given biomarker. Flow showed overall very little improvements in 2D cultures but a slight improvement in 3D cultures suggesting that high density cell culture may benefit from flow. In conclusion, the gains of perfusion are relatively modest, larger gains are linked to specific biomarkers in certain cell types.
Collapse
Affiliation(s)
- Martin Dufva
- Department of Health Technology, Technical University of Denmark, 2800, Kgs Lyngby, Denmark.
| |
Collapse
|
10
|
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
|
11
|
Gravity-driven microfluidic device placed on a slow-tilting table enables constant unidirectional perfusion culture of human induced pluripotent stem cells. J Biosci Bioeng 2023; 135:151-159. [PMID: 36586792 DOI: 10.1016/j.jbiosc.2022.11.007] [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: 03/14/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/30/2022]
Abstract
Gravity-driven microfluidics, which utilizes gravity force to drive liquid flow, offers portability and multi-condition setting flexibility because they do not require pumps or connection tubes to drive the flow. However, because the flow rate decreases with time in gravity-driven microfluidics, it is not suitable for stem cell experiments, which require long-term (at least a day) stability. In this study, gravity-driven microfluidics and a slow-tilting table were developed to culture cells under constant unidirectional perfusion. The microfluidic device was placed on a slow-tilting table, which tilts unidirectionally at a rate of approximately 7° per day to compensate for the reduction in the flow rate. Computational simulations showed that the pulsation of the flow arising from the stepwise movement of the table was less than 0.2%, and the flow was laminar. Hydrophilization of the tanks increased the flow rate, which is consistent with the theoretical values. We showed that vitronectin is better than laminin 511 fragments as a coating material for adhering human induced pluripotent stem cells on a microchamber made of polydimethylsiloxane, and succeeded in culturing the cells for 3 days. It is believed that the system offers easy-to-use cell culture tools, such as conventional multiwell culture vessels, and enables the control of the cell microenvironment.
Collapse
|
12
|
Singh NK, Kim JY, Lee JY, Lee H, Gao G, Jang J, Kim YK, Cho DW. Coaxial cell printing of a human glomerular model: an in vitroglomerular filtration barrier and its pathophysiology. Biofabrication 2023; 15. [PMID: 36538823 DOI: 10.1088/1758-5090/acad2c] [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: 08/07/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Much effort has been expended in emulating the kidney's glomerular unit because of its limitless potential in the field of drug screening and nephrotoxicity testing in clinics. Herein, we fabricate a functional bilayer glomerular microvessel-on-a-chip that recapitulates the specific arrangement of the glomerular endothelial cell, podocyte layers, and the intervening glomerular basement membrane (GBM) in a single step. Our perfusable chip allows for the co-culture of monolayer glomerular endothelium and podocyte epithelium, which display mature functional markers of glomerular cells, and their proper interactions produce GBM proteins, which are the major components of the GBMin vivo. Furthermore, we test the selective permeability capacity, a representative hallmark function of the glomerular filtration barrier. Lastly, we evaluate the response of our glomerular model to Adriamycin- and hyperglycemia-induced injury to evaluate its applicability for drug screening and glomerular disease modeling.
Collapse
Affiliation(s)
- Narendra K Singh
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Division of Biomaterials and Biomechanics, School of Dentistry, Oregon Health and Science University (OHSU), Portland, OR 97201, United States of America.,Cancer Early Detection Advanced Research Center (CEDAR), OHSU-Knight Cancer Institute, Portland, OR 97201, United States of America
| | - Jae Yun Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jae Yeon Lee
- Department of Companion Animal Health, Daegu Haany University, Gyeongsan, Republic of Korea
| | - Hyungseok Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon, Republic of Korea.,Interdisciplinary Program in Biohealth-Machinery Convergence Engineering, Kangwon National University (KNU), Chuncheon, Republic of Korea
| | - Ge Gao
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| | - Yong Kyun Kim
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Internal Medicine, College of Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon, Republic of Korea.,POSTECH-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Shuchat S, Yossifon G, Huleihel M. Perfusion in Organ-on-Chip Models and Its Applicability to the Replication of Spermatogenesis In Vitro. Int J Mol Sci 2022; 23:5402. [PMID: 35628214 PMCID: PMC9141186 DOI: 10.3390/ijms23105402] [Citation(s) in RCA: 3] [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: 03/31/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
Organ/organoid-on-a-chip (OoC) technologies aim to replicate aspects of the in vivo environment in vitro, at the scale of microns. Mimicking the spatial in vivo structure is important and can provide a deeper understanding of the cell-cell interactions and the mechanisms that lead to normal/abnormal function of a given organ. It is also important for disease models and drug/toxin testing. Incorporating active fluid flow in chip models enables many more possibilities. Active flow can provide physical cues, improve intercellular communication, and allow for the dynamic control of the environment, by enabling the efficient introduction of biological factors, drugs, or toxins. All of this is in addition to the fundamental role of flow in supplying nutrition and removing waste metabolites. This review presents an overview of the different types of fluid flow and how they are incorporated in various OoC models. The review then describes various methods and techniques of incorporating perfusion networks into OoC models, including self-assembly, bioprinting techniques, and utilizing sacrificial gels. The second part of the review focuses on the replication of spermatogenesis in vitro; the complex process whereby spermatogonial stem cells differentiate into mature sperm. A general overview is given of the various approaches that have been used. The few studies that incorporated microfluidics or vasculature are also described. Finally, a future perspective is given on elements from perfusion-based models that are currently used in models of other organs and can be applied to the field of in vitro spermatogenesis. For example, adopting tubular blood vessel models to mimic the morphology of the seminiferous tubules and incorporating vasculature in testis-on-a-chip models. Improving these models would improve our understanding of the process of spermatogenesis. It may also potentially provide novel therapeutic strategies for pre-pubertal cancer patients who need aggressive chemotherapy that can render them sterile, as well asfor a subset of non-obstructive azoospermic patients with maturation arrest, whose testes do not produce sperm but still contain some of the progenitor cells.
Collapse
Affiliation(s)
- Sholom Shuchat
- Faculty of Mechanical Engineering, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (S.S.); (G.Y.)
| | - Gilad Yossifon
- Faculty of Mechanical Engineering, Technion–Israel Institute of Technology, Haifa 3200003, Israel; (S.S.); (G.Y.)
- School of Mechanical Engineering, University of Tel Aviv, Tel Aviv 6997801, Israel
| | - Mahmoud Huleihel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Science, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- The Center of Advanced Research and Education in Reproduction (CARER), Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 8410501, Israel
| |
Collapse
|
14
|
De Stefano P, Bianchi E, Dubini G. The impact of microfluidics in high-throughput drug-screening applications. BIOMICROFLUIDICS 2022; 16:031501. [PMID: 35646223 PMCID: PMC9142169 DOI: 10.1063/5.0087294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/02/2022] [Indexed: 05/05/2023]
Abstract
Drug discovery is an expensive and lengthy process. Among the different phases, drug discovery and preclinical trials play an important role as only 5-10 of all drugs that begin preclinical tests proceed to clinical trials. Indeed, current high-throughput screening technologies are very expensive, as they are unable to dispense small liquid volumes in an accurate and quick way. Moreover, despite being simple and fast, drug screening assays are usually performed under static conditions, thus failing to recapitulate tissue-specific architecture and biomechanical cues present in vivo even in the case of 3D models. On the contrary, microfluidics might offer a more rapid and cost-effective alternative. Although considered incompatible with high-throughput systems for years, technological advancements have demonstrated how this gap is rapidly reducing. In this Review, we want to further outline the role of microfluidics in high-throughput drug screening applications by looking at the multiple strategies for cell seeding, compartmentalization, continuous flow, stimuli administration (e.g., drug gradients or shear stresses), and single-cell analyses.
Collapse
Affiliation(s)
- Paola De Stefano
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Elena Bianchi
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Gabriele Dubini
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| |
Collapse
|
15
|
Riddle RB, Jennbacken K, Hansson KM, Harper MT. Endothelial inflammation and neutrophil transmigration are modulated by extracellular matrix composition in an inflammation-on-a-chip model. Sci Rep 2022; 12:6855. [PMID: 35477984 PMCID: PMC9046410 DOI: 10.1038/s41598-022-10849-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/11/2022] [Indexed: 12/20/2022] Open
Abstract
Inflammatory diseases are often characterised by excessive neutrophil infiltration from the blood stream to the site of inflammation, which damages healthy tissue and prevents resolution of inflammation. Development of anti-inflammatory drugs is hindered by lack of in vitro and in vivo models which accurately represent the disease microenvironment. In this study, we used the OrganoPlate to develop a humanized 3D in vitro inflammation-on-a-chip model to recapitulate neutrophil transmigration across the endothelium and subsequent migration through the extracellular matrix (ECM). Human umbilical vein endothelial cells formed confluent vessels against collagen I and geltrex mix, a mix of basement membrane extract and collagen I. TNF-α-stimulation of vessels upregulated inflammatory cytokine expression and promoted neutrophil transmigration. Intriguingly, major differences were found depending on the composition of the ECM. Neutrophils transmigrated in higher number and further in geltrex mix than collagen I, and did not require an N-formyl-methionyl-leucyl-phenylalanine (fMLP) gradient for transmigration. Inhibition of neutrophil proteases inhibited neutrophil transmigration on geltrex mix, but not collagen I. These findings highlight the important role of the ECM in determining cell phenotype and response to inhibitors. Future work could adapt the ECM composition for individual diseases, producing accurate models for drug development.
Collapse
Affiliation(s)
- Rebecca B Riddle
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Kenny M Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
16
|
Fallon ME, Mathews R, Hinds MT. In Vitro Flow Chamber Design for the Study of Endothelial Cell (Patho)Physiology. J Biomech Eng 2022; 144:020801. [PMID: 34254640 PMCID: PMC8628846 DOI: 10.1115/1.4051765] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 07/06/2021] [Indexed: 02/03/2023]
Abstract
In the native vasculature, flowing blood produces a frictional force on vessel walls that affects endothelial cell function and phenotype. In the arterial system, the vasculature's local geometry directly influences variations in flow profiles and shear stress magnitudes. Straight arterial sections with pulsatile shear stress have been shown to promote an athero-protective endothelial phenotype. Conversely, areas with more complex geometry, such as arterial bifurcations and branch points with disturbed flow patterns and lower, oscillatory shear stress, typically lead to endothelial dysfunction and the pathogenesis of cardiovascular diseases. Many studies have investigated the regulation of endothelial responses to various shear stress environments. Importantly, the accurate in vitro simulation of in vivo hemodynamics is critical to the deeper understanding of mechanotransduction through the proper design and use of flow chamber devices. In this review, we describe several flow chamber apparatuses and their fluid mechanics design parameters, including parallel-plate flow chambers, cone-and-plate devices, and microfluidic devices. In addition, chamber-specific design criteria and relevant equations are defined in detail for the accurate simulation of shear stress environments to study endothelial cell responses.
Collapse
Affiliation(s)
- Meghan E. Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| | - Rick Mathews
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, 3303 S Bond Ave CH13B, Portland, OR 97239
| |
Collapse
|
17
|
Lee SY, Kim D, Lee SH, Sung JH. Microtechnology-based in vitro models: Mimicking liver function and pathophysiology. APL Bioeng 2021; 5:041505. [PMID: 34703969 PMCID: PMC8520487 DOI: 10.1063/5.0061896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
The liver plays important roles in drug metabolism and homeostasis. The metabolism and biotransformation can not only affect the efficacy of drugs but also result in hepatotoxicity and drug-induced liver injury. Understanding the complex physiology of the liver and the pathogenetic mechanisms of liver diseases is essential for drug development. Conventional in vitro models have limitations in the ability to predict drug effects, due to the lack of physiological relevance. Recently, the liver-on-a-chip platform has been developed to reproduce the microarchitecture and in vivo environment of the liver. These efforts have improved the physiological relevance of the liver tissue used in the platform and have demonstrated its applicability to drug screening and disease models. In this review, we summarize the recent development of liver-on-a-chip models that closely mimic the in vivo liver environments and liver diseases.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| | - Donghyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, South Korea
| | - Seung Hwan Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, South Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| |
Collapse
|
18
|
Development of an Aged Full-Thickness Skin Model Using Flexible Skin-on-a-Chip Subjected to Mechanical Stimulus Reflecting the Circadian Rhythm. Int J Mol Sci 2021; 22:ijms222312788. [PMID: 34884594 PMCID: PMC8657468 DOI: 10.3390/ijms222312788] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022] Open
Abstract
The skin is subject to both intrinsic aging caused by metabolic processes in the body and extrinsic aging caused by exposure to environmental factors. Intrinsic aging is an important obstacle to in vitro experimentation as its long-term progression is difficult to replicate. Here, we accelerated aging of a full-thickness skin equivalent by applying periodic mechanical stimulation, replicating the circadian rhythm for 28 days. This aging skin model was developed by culturing a full-thickness, three-dimensional skin equivalent with human fibroblasts and keratinocytes to produce flexible skin-on-a-chip. Accelerated aging associated with periodic compressive stress was evidenced by reductions in the epidermal layer thickness, contraction rate, and secretion of Myb. Increases in β-galactosidase gene expression and secretion of reactive oxygen species and transforming growth factor-β1 were also observed. This in vitro aging skin model is expected to greatly accelerate drug development for skin diseases and cosmetics that cannot be tested on animals.
Collapse
|
19
|
Lee Y, Kim MH, Alves DR, Kim S, Lee LP, Sung JH, Park S. Gut-Kidney Axis on Chip for Studying Effects of Antibiotics on Risk of Hemolytic Uremic Syndrome by Shiga Toxin-Producing Escherichia coli. Toxins (Basel) 2021; 13:toxins13110775. [PMID: 34822559 PMCID: PMC8622205 DOI: 10.3390/toxins13110775] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/23/2021] [Accepted: 10/30/2021] [Indexed: 12/30/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) infects humans by colonizing the large intestine, and causes kidney damage by secreting Shiga toxins (Stxs). The increased secretion of Shiga toxin 2 (Stx2) by some antibiotics, such as ciprofloxacin (CIP), increases the risk of hemolytic–uremic syndrome (HUS), which can be life-threatening. However, previous studies evaluating this relationship have been conflicting, owing to the low frequency of EHEC infection, very small number of patients, and lack of an appropriate animal model. In this study, we developed gut–kidney axis (GKA) on chip for co-culturing gut (Caco-2) and kidney (HKC-8) cells, and observed both STEC O157:H7 (O157) infection and Stx intoxication in the gut and kidney cells on the chip, respectively. Without any antibiotic treatment, O157 killed both gut and kidney cells in GKA on the chip. CIP treatment reduced O157 infection in the gut cells, but increased Stx2-induced damage in the kidney cells, whereas the gentamycin treatment reduced both O157 infection in the gut cells and Stx2-induced damage in the kidney cells. This is the first report to recapitulate a clinically relevant situation, i.e., that CIP treatment causes more damage than gentamicin treatment. These results suggest that GKA on chip is very useful for simultaneous observation of O157 infections and Stx2 poisoning in gut and kidney cells, making it suitable for studying the effects of antibiotics on the risk of HUS.
Collapse
Affiliation(s)
- Yugyeong Lee
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
| | - David Rodrigues Alves
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, 1362035 Lisboa, Portugal
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Luke P. Lee
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Korea
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| | - Sungsu Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea; (M.-H.K.); (D.R.A.)
- Institute of Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University (SKKU), Suwon 16419, Korea;
- Correspondence: (J.H.S.); (S.P.); Tel.: +82-2-320-3067 (J.H.S.); +82-31-290-7431 (S.P.)
| |
Collapse
|
20
|
Schneider S, Bubeck M, Rogal J, Weener HJ, Rojas C, Weiss M, Heymann M, van der Meer AD, Loskill P. Peristaltic on-chip pump for tunable media circulation and whole blood perfusion in PDMS-free organ-on-chip and Organ-Disc systems. LAB ON A CHIP 2021; 21:3963-3978. [PMID: 34636813 DOI: 10.1039/d1lc00494h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Organ-on-chip (OoC) systems have become a promising tool for personalized medicine and drug development with advantages over conventional animal models and cell assays. However, the utility of OoCs in industrial settings is still limited, as external pumps and tubing for on-chip fluid transport are dependent on error-prone, manual handling. Here, we present an on-chip pump for OoC and Organ-Disc systems, to perfuse media without external pumps or tubing. Peristaltic pumping is implemented through periodic compression of a flexible pump layer. The disc-shaped, microfluidic module contains four independent systems, each lined with endothelial cells cultured under defined, peristaltic perfusion. Both cell viability and functionality were maintained over several days shown by supernatant analysis and immunostaining. Integrated, on-disc perfusion was further used for cytokine-induced cell activation with physiologic cell responses and for whole blood perfusion assays, both demonstrating the versatility of our system for OoC applications.
Collapse
Affiliation(s)
- Stefan Schneider
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Marvin Bubeck
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | - Julia Rogal
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Department of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany.
| | - Huub J Weener
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Applied Stem Cell Technologies, University of Twente, Enschede, The Netherlands
| | - Cristhian Rojas
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department of Women's Health, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Michael Heymann
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| | | | - Peter Loskill
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Department of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany.
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- 3R-Center for in vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
21
|
Jones CG, Huang T, Chung JH, Chen C. 3D-Printed, Modular, and Parallelized Microfluidic System with Customizable Scaffold Integration to Investigate the Roles of Basement Membrane Topography on Endothelial Cells. ACS Biomater Sci Eng 2021; 7:1600-1607. [PMID: 33545000 DOI: 10.1021/acsbiomaterials.0c01752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Because dysfunctions of endothelial cells are involved in many pathologies, in vitro endothelial cell models for pathophysiological and pharmaceutical studies have been a valuable research tool. Although numerous microfluidic-based endothelial models have been reported, they had the cells cultured on a flat surface without considering the possible three-dimensional (3D) structure of the native extracellular matrix (ECM). Endothelial cells rest on the basement membrane in vivo, which contains an aligned microfibrous topography. To better understand and model the cells, it is necessary to know if and how the fibrous topography can affect endothelial functions. With conventional fully integrated microfluidic apparatus, it is difficult to include additional topographies in a microchannel. Therefore, we developed a modular microfluidic system by 3D-printing and electrospinning, which enabled easy integration and switching of desired ECM topographies. Also, with standardized designs, the system allowed for high flow rates up to 4000 μL/min, which encompassed the full shear stress range for endothelial studies. We found that the aligned fibrous topography on the ECM altered arginine metabolism in endothelial cells and thus increased nitric oxide production. There has not been an endothelial model like this, and the new knowledge generated thereby lays a groundwork for future endothelial research and modeling.
Collapse
Affiliation(s)
- Curtis G Jones
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| | - Tianjiao Huang
- Laboratory of Obesity and Aging Research, Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, Maryland 20892, United States
| | - Jay H Chung
- Laboratory of Obesity and Aging Research, Cardiovascular Branch, National Heart Lung and Blood Institute, Bethesda, Maryland 20892, United States
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
22
|
Akther F, Yakob SB, Nguyen NT, Ta HT. Surface Modification Techniques for Endothelial Cell Seeding in PDMS Microfluidic Devices. BIOSENSORS 2020; 10:E182. [PMID: 33228050 PMCID: PMC7699314 DOI: 10.3390/bios10110182] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
Microfluidic lab-on-a-chip cell culture techniques have been gaining popularity by offering the possibility of reducing the amount of samples and reagents and greater control over cellular microenvironment. Polydimethylsiloxane (PDMS) is the commonly used polymer for microfluidic cell culture devices because of the cheap and easy fabrication techniques, non-toxicity, biocompatibility, high gas permeability, and optical transparency. However, the intrinsic hydrophobic nature of PDMS makes cell seeding challenging when applied on PDMS surface. The hydrophobicity of the PDMS surface also allows the non-specific absorption/adsorption of small molecules and biomolecules that might affect the cellular behaviour and functions. Hydrophilic modification of PDMS surface is indispensable for successful cell seeding. This review collates different techniques with their advantages and disadvantages that have been used to improve PDMS hydrophilicity to facilitate endothelial cells seeding in PDMS devices.
Collapse
Affiliation(s)
- Fahima Akther
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Shazwani Binte Yakob
- School of Pharmacy, the University of Queensland, Brisbane, QLD 4102, Australia;
| | - Nam-Trung Nguyen
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
| | - Hang T. Ta
- Australian Institute for Bioengineering and Nanotechnology, the University of Queensland, Brisbane, QLD 4072, Australia;
- Queensland Micro-and Nanotechnology Centre, Griffith University, Brisbane, QLD 4111, Australia;
- School of Environment and Science, Griffith University, Brisbane, QLD 4111, Australia
| |
Collapse
|
23
|
Kim J, Kim K, Sung GY. Coenzyme Q10 Efficacy Test for Human Skin Equivalents Using a Pumpless Skin-On-A-Chip System. Int J Mol Sci 2020; 21:ijms21228475. [PMID: 33187177 PMCID: PMC7697574 DOI: 10.3390/ijms21228475] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/31/2020] [Accepted: 11/08/2020] [Indexed: 11/29/2022] Open
Abstract
A human skin equivalent (HSE) composed of the epidermis and dermis is cultured using a pumpless skin-on-a-chip system to supply cultures the desired flow rate using gravity flow without a pump or an external tube connection. Coenzyme Q10 efficacy is tested by adjusting its concentration, as it is known to have anti-aging and antioxidant effects in culture solutions. The relationship between the contraction rate of a full-thickness human skin equivalent and secreted transforming growth factor (TGF) β-1 is analyzed via enzyme-linked immunosorbent assay (ELISA). Following hematoxylin and eosin (H&E) staining, an image of the skin equivalent is analyzed to measure the epidermal layer’s thickness. The cell density and differentiation of the dermis layer are investigated. Gene and protein expression in the dermal and epidermal layers are quantitatively analyzed using quantitative real time polymerase chain reaction (qPCR) and immunohistochemical staining. As the coenzyme Q10 treatment concentration increased, the number of cells per unit area and the thickness of the epidermal layer increased, the expression level of filaggrin increased, and the contraction rate of full-thickness HSE was proportional to the amount of TGF β-1 secreted.
Collapse
Affiliation(s)
- Jisue Kim
- Cooperative Course of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (J.K.); (K.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Kyunghee Kim
- Cooperative Course of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (J.K.); (K.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Gun Yong Sung
- Cooperative Course of Nano-Medical Device Engineering, Graduate School, Hallym University, Chuncheon 24252, Korea; (J.K.); (K.K.)
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
- Major in Materials Science and Engineering, School of Future Convergence, Hallym University, Chuncheon 24252, Korea
- Correspondence:
| |
Collapse
|
24
|
Kwak BS, Jin SP, Kim SJ, Kim EJ, Chung JH, Sung JH. Microfluidic skin chip with vasculature for recapitulating the immune response of the skin tissue. Biotechnol Bioeng 2020; 117:1853-1863. [PMID: 32100875 DOI: 10.1002/bit.27320] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/16/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
Abstract
There is a considerable need for cell-based in vitro skin models for studying dermatological diseases and testing cosmetic products, but current in vitro skin models lack physiological relevance compared to human skin tissue. For example, many dermatological disorders involve complex immune responses, but current skin models are not capable of recapitulating the phenomena. Previously, we reported development of a microfluidic skin chip with a vessel structure and vascular endothelial cells. In this study, we cocultured dermal fibroblasts and keratinocytes with vascular endothelial cells, human umbilical vascular endothelial cells. We verified the formation of a vascular endothelium in the presence of the dermis and epidermis layers by examining the expression of tissue-specific markers. As the vascular endothelium plays a critical role in the migration of leukocytes to inflammation sites, we incorporated leukocytes in the circulating media and attempted to mimic the migration of neutrophils in response to external stimuli. Increased secretion of cytokines and migration of neutrophils was observed when the skin chip was exposed to ultraviolet irradiation, showing that the microfluidic skin chip may be useful for studying the immune response of the human tissue.
Collapse
Affiliation(s)
- Bong Shin Kwak
- Department of Chemical Engineering, Hongik University, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Su Jung Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Joo Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Republic of Korea
| |
Collapse
|
25
|
Jeon HM, Kim K, Choi KC, Sung GY. Side-effect test of sorafenib using 3-D skin equivalent based on microfluidic skin-on-a-chip. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2019.09.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
26
|
de Mello CPP, Rumsey J, Slaughter V, Hickman JJ. A human-on-a-chip approach to tackling rare diseases. Drug Discov Today 2019; 24:2139-2151. [PMID: 31412288 PMCID: PMC6856435 DOI: 10.1016/j.drudis.2019.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/18/2019] [Accepted: 08/05/2019] [Indexed: 12/20/2022]
Abstract
Drug development for rare diseases, classified as diseases with a prevalence of < 200 000 patients, is limited by the high cost of research and low target population. Owing to a lack of representative disease models, research has been challenging for orphan drugs. Human-on-a-chip (HoaC) technology, which models human tissues in interconnected in vitro microfluidic devices, has the potential to lower the cost of preclinical studies and increase the rate of drug approval by introducing human phenotypic models early in the drug discovery process. Advances in HoaC technology can drive a new approach to rare disease research and orphan drug development.
Collapse
Affiliation(s)
| | | | - Victoria Slaughter
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA; Hesperos, Inc., Orlando, FL 32826, USA.
| |
Collapse
|
27
|
Ragelle H, Goncalves A, Kustermann S, Antonetti DA, Jayagopal A. Organ-On-A-Chip Technologies for Advanced Blood-Retinal Barrier Models. J Ocul Pharmacol Ther 2019; 36:30-41. [PMID: 31140899 PMCID: PMC6985766 DOI: 10.1089/jop.2019.0017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
The blood-retinal barrier (BRB) protects the retina by maintaining an adequate microenvironment for neuronal function. Alterations of the junctional complex of the BRB and consequent BRB breakdown in disease contribute to a loss of neuronal signaling and vision loss. As new therapeutics are being developed to prevent or restore barrier function, it is critical to implement physiologically relevant in vitro models that recapitulate the important features of barrier biology to improve disease modeling, target validation, and toxicity assessment. New directions in organ-on-a-chip technology are enabling more sophisticated 3-dimensional models with flow, multicellularity, and control over microenvironmental properties. By capturing additional biological complexity, organs-on-chip can help approach actual tissue organization and function and offer additional tools to model and study disease compared with traditional 2-dimensional cell culture. This review describes the current state of barrier biology and barrier function in ocular diseases, describes recent advances in organ-on-a-chip design for modeling the BRB, and discusses the potential of such models for ophthalmic drug discovery and development.
Collapse
Affiliation(s)
- Héloïse Ragelle
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Stefan Kustermann
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - David A. Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Ashwath Jayagopal
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
28
|
Renggli K, Rousset N, Lohasz C, Nguyen OTP, Hierlemann A. Integrated Microphysiological Systems: Transferable Organ Models and Recirculating Flow. ADVANCED BIOSYSTEMS 2019; 3:e1900018. [PMID: 32627410 PMCID: PMC7610576 DOI: 10.1002/adbi.201900018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.
Collapse
Affiliation(s)
- Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | | | |
Collapse
|
29
|
Sung JH, Wang YI, Narasimhan Sriram N, Jackson M, Long C, Hickman JJ, Shuler ML. Recent Advances in Body-on-a-Chip Systems. Anal Chem 2019; 91:330-351. [PMID: 30472828 PMCID: PMC6687466 DOI: 10.1021/acs.analchem.8b05293] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering , Hongik University , Seoul , 04066 , Republic of Korea
| | - Ying I Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering , Cornell University , Ithaca , New York 14853 , United States
| | | | - Max Jackson
- Hesperos, Inc. Orlando , Florida 32836 , United States
| | | | - James J Hickman
- Hesperos, Inc. Orlando , Florida 32836 , United States
- NanoScience Technology Center , University of Central Florida , Orlando , Florida 32828 , United States
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering , Cornell University , Ithaca , New York 14853 , United States
- Hesperos, Inc. Orlando , Florida 32836 , United States
- Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , New York 14853 , United States
| |
Collapse
|