201
|
Poletti M, Arnauts K, Ferrante M, Korcsmaros T. Organoid-based Models to Study the Role of Host-microbiota Interactions in IBD. J Crohns Colitis 2020; 15:1222-1235. [PMID: 33341879 PMCID: PMC8256633 DOI: 10.1093/ecco-jcc/jjaa257] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gut microbiota appears to play a central role in health, and alterations in the gut microbiota are observed in both forms of inflammatory bowel disease [IBD], namely Crohn's disease and ulcerative colitis. Yet, the mechanisms behind host-microbiota interactions in IBD, especially at the intestinal epithelial cell level, are not yet fully understood. Dissecting the role of host-microbiota interactions in disease onset and progression is pivotal, and requires representative models mimicking the gastrointestinal ecosystem, including the intestinal epithelium, the gut microbiota, and immune cells. New advancements in organoid microfluidics technology are facilitating the study of IBD-related microbial-epithelial cross-talk, and the discovery of novel microbial therapies. Here, we review different organoid-based ex vivo models that are currently available, and benchmark their suitability and limitations for specific research questions. Organoid applications, such as patient-derived organoid biobanks for microbial screening and 'omics technologies, are discussed, highlighting their potential to gain better mechanistic insights into disease mechanisms and eventually allow personalised medicine.
Collapse
Affiliation(s)
- Martina Poletti
- Earlham Institute, Norwich Research Park, Norwich, UK,Quadram Institute, Norwich Research Park, Norwich, UK
| | - Kaline Arnauts
- Department of Chronic Diseases, Metabolism and Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium,Department of Development and Regeneration, Stem Cell Institute Leuven [SCIL], KU Leuven, Leuven, Belgium
| | - Marc Ferrante
- Department of Chronic Diseases, Metabolism and Ageing [CHROMETA], Translational Research Center for Gastrointestinal Disorders [TARGID], KU Leuven, Leuven, Belgium,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium,Corresponding author: Marc Ferrante, MD, PhD, Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium. Tel.: +32 16 344225;
| | - Tamas Korcsmaros
- Earlham Institute, Norwich Research Park, Norwich, UK,Quadram Institute, Norwich Research Park, Norwich, UK
| |
Collapse
|
202
|
|
203
|
Beaurivage C, Kanapeckaite A, Loomans C, Erdmann KS, Stallen J, Janssen RAJ. Development of a human primary gut-on-a-chip to model inflammatory processes. Sci Rep 2020; 10:21475. [PMID: 33293676 PMCID: PMC7722760 DOI: 10.1038/s41598-020-78359-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a complex multi-factorial disease for which physiologically relevant in vitro models are lacking. Existing models are often a compromise between biological relevance and scalability. Here, we integrated intestinal epithelial cells (IEC) derived from human intestinal organoids with monocyte-derived macrophages, in a gut-on-a-chip platform to model the human intestine and key aspects of IBD. The microfluidic culture of IEC lead to an increased polarization and differentiation state that closely resembled the expression profile of human colon in vivo. Activation of the model resulted in the polarized secretion of CXCL10, IL-8 and CCL-20 by IEC and could efficiently be prevented by TPCA-1 exposure. Importantly, upregulated gene expression by the inflammatory trigger correlated with dysregulated pathways in IBD patients. Finally, integration of activated macrophages offers a first-step towards a multi-factorial amenable IBD platform that could be scaled up to assess compound efficacy at early stages of drug development or in personalized medicine.
Collapse
Affiliation(s)
- Claudia Beaurivage
- Galapagos BV, Leiden, South Holland, 2333CL, The Netherlands
- Department of Biomedical Science, Faculty of Science, University of Sheffield, Sheffield, S10 2TN, South Yorkshire, UK
| | | | - Cindy Loomans
- Galapagos BV, Leiden, South Holland, 2333CL, The Netherlands
| | - Kai S Erdmann
- Department of Biomedical Science, Faculty of Science, University of Sheffield, Sheffield, S10 2TN, South Yorkshire, UK
| | - Jan Stallen
- Galapagos BV, Leiden, South Holland, 2333CL, The Netherlands
| | | |
Collapse
|
204
|
Onfroy-Roy L, Hamel D, Foncy J, Malaquin L, Ferrand A. Extracellular Matrix Mechanical Properties and Regulation of the Intestinal Stem Cells: When Mechanics Control Fate. Cells 2020; 9:cells9122629. [PMID: 33297478 PMCID: PMC7762382 DOI: 10.3390/cells9122629] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Intestinal stem cells (ISC) are crucial players in colon epithelium physiology. The accurate control of their auto-renewal, proliferation and differentiation capacities provides a constant flow of regeneration, maintaining the epithelial intestinal barrier integrity. Under stress conditions, colon epithelium homeostasis in disrupted, evolving towards pathologies such as inflammatory bowel diseases or colorectal cancer. A specific environment, namely the ISC niche constituted by the surrounding mesenchymal stem cells, the factors they secrete and the extracellular matrix (ECM), tightly controls ISC homeostasis. Colon ECM exerts physical constraint on the enclosed stem cells through peculiar topography, stiffness and deformability. However, little is known on the molecular and cellular events involved in ECM regulation of the ISC phenotype and fate. To address this question, combining accurately reproduced colon ECM mechanical parameters to primary ISC cultures such as organoids is an appropriated approach. Here, we review colon ECM physical properties at physiological and pathological states and their bioengineered in vitro reproduction applications to ISC studies.
Collapse
Affiliation(s)
- Lauriane Onfroy-Roy
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- Correspondence: (L.O.-R.); (A.F.); Tel.: +33-5-62-744-522 (A.F.)
| | - Dimitri Hamel
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Julie Foncy
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Laurent Malaquin
- LAAS-CNRS, Université de Toulouse, CNRS, 31400 Toulouse, France; (J.F.); (L.M.)
| | - Audrey Ferrand
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, 31024 Toulouse, France;
- Correspondence: (L.O.-R.); (A.F.); Tel.: +33-5-62-744-522 (A.F.)
| |
Collapse
|
205
|
Yuan L, de Haan P, Peterson BW, de Jong ED, Verpoorte E, van der Mei HC, Busscher HJ. Visualization of Bacterial Colonization and Cellular Layers in a Gut-on-a-Chip System Using Optical Coherence Tomography. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:1211-1219. [PMID: 33107427 DOI: 10.1017/s143192762002454x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Imaging of cellular layers in a gut-on-a-chip system has been confined to two-dimensional (2D)-imaging through conventional light microscopy and confocal laser scanning microscopy (CLSM) yielding three-dimensional- and 2D-cross-sectional reconstructions. However, CLSM requires staining and is unsuitable for longitudinal visualization. Here, we compare merits of optical coherence tomography (OCT) with those of CLSM and light microscopy for visualization of intestinal epithelial layers during protection by a probiotic Bifidobacterium breve strain and a simultaneous pathogen challenge by an Escherichia coli strain. OCT cross-sectional images yielded film thicknesses that coincided with end-point thicknesses derived from cross-sectional CLSM images. Light microscopy on histological sections of epithelial layers at the end-point yielded smaller layer thicknesses than OCT and CLSM. Protective effects of B. breve adhering to an epithelial layer against an E. coli challenge included the preservation of layer thickness and membrane surface coverage by epithelial cells. OCT does not require staining or sectioning, making OCT suitable for longitudinal visualization of biological films, but as a drawback, OCT does not allow an epithelial layer to be distinguished from bacterial biofilms adhering to it. Thus, OCT is ideal to longitudinally evaluate epithelial layers under probiotic protection and pathogen challenges, but proper image interpretation requires the application of a second method at the end-point to distinguish bacterial and epithelial films.
Collapse
Affiliation(s)
- Lu Yuan
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, 9713 AVGroningen, The Netherlands
| | - Pim de Haan
- University of Groningen, Groningen Research Institute of Pharmacy, Pharmaceutical Analysis, 9713 AVGroningen, The Netherlands
- TI-COAST, 1098 XHAmsterdam, The Netherlands
| | - Brandon W Peterson
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, 9713 AVGroningen, The Netherlands
| | - Ed D de Jong
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, 9713 AVGroningen, The Netherlands
| | - Elisabeth Verpoorte
- University of Groningen, Groningen Research Institute of Pharmacy, Pharmaceutical Analysis, 9713 AVGroningen, The Netherlands
| | - Henny C van der Mei
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, 9713 AVGroningen, The Netherlands
| | - Henk J Busscher
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, 9713 AVGroningen, The Netherlands
| |
Collapse
|
206
|
Salah E, Abouelfetouh MM, Pan Y, Chen D, Xie S. Solid lipid nanoparticles for enhanced oral absorption: A review. Colloids Surf B Biointerfaces 2020; 196:111305. [DOI: 10.1016/j.colsurfb.2020.111305] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 12/26/2022]
|
207
|
Pasman T, Baptista D, van Riet S, Truckenmüller RK, Hiemstra PS, Rottier RJ, Stamatialis D, Poot AA. Development of Porous and Flexible PTMC Membranes for In Vitro Organ Models Fabricated by Evaporation-Induced Phase Separation. MEMBRANES 2020; 10:E330. [PMID: 33167539 PMCID: PMC7694515 DOI: 10.3390/membranes10110330] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 11/16/2022]
Abstract
Polymeric membranes are widely applied in biomedical applications, including in vitro organ models. In such models, they are mostly used as supports on which cells are cultured to create functional tissue units of the desired organ. To this end, the membrane properties, e.g., morphology and porosity, should match the tissue properties. Organ models of dynamic (barrier) tissues, e.g., lung, require flexible, elastic and porous membranes. Thus, membranes based on poly (dimethyl siloxane) (PDMS) are often applied, which are flexible and elastic. However, PDMS has low cell adhesive properties and displays small molecule ad- and absorption. Furthermore, the introduction of porosity in these membranes requires elaborate methods. In this work, we aim to develop porous membranes for organ models based on poly(trimethylene carbonate) (PTMC): a flexible polymer with good cell adhesive properties which has been used for tissue engineering scaffolds, but not in in vitro organ models. For developing these membranes, we applied evaporation-induced phase separation (EIPS), a new method in this field based on solvent evaporation initiating phase separation, followed by membrane photo-crosslinking. We optimised various processing variables for obtaining form-stable PTMC membranes with average pore sizes between 5 to 8 µm and water permeance in the microfiltration range (17,000-41,000 L/m2/h/bar). Importantly, the membranes are flexible and are suitable for implementation in in vitro organ models.
Collapse
Affiliation(s)
- Thijs Pasman
- Department of Biomaterials Science and Technology, Technical Medical (TechMed) Centre, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands; (T.P.); (D.S.)
| | - Danielle Baptista
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.B.); (R.K.T.)
| | - Sander van Riet
- Department of Pulmonology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands; (S.v.R.); (P.S.H.)
| | - Roman K. Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (D.B.); (R.K.T.)
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Centre, 2300 RC Leiden, The Netherlands; (S.v.R.); (P.S.H.)
| | - Robbert J. Rottier
- Department of Pediatric Surgery, Erasmus MC-Sophia Children’s Hospital, 3000 CB Rotterdam, The Netherlands;
| | - Dimitrios Stamatialis
- Department of Biomaterials Science and Technology, Technical Medical (TechMed) Centre, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands; (T.P.); (D.S.)
| | - André A. Poot
- Department of Biomaterials Science and Technology, Technical Medical (TechMed) Centre, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands; (T.P.); (D.S.)
| |
Collapse
|
208
|
Ingber DE. Is it Time for Reviewer 3 to Request Human Organ Chip Experiments Instead of Animal Validation Studies? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002030. [PMID: 33240763 PMCID: PMC7675190 DOI: 10.1002/advs.202002030] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/22/2020] [Indexed: 05/08/2023]
Abstract
For the past century, experimental data obtained from animal studies have been required by reviewers of scientific articles and grant applications to validate the physiological relevance of in vitro results. At the same time, pharmaceutical researchers and regulatory agencies recognize that results from preclinical animal models frequently fail to predict drug responses in humans. This Progress Report reviews recent advances in human organ-on-a-chip (Organ Chip) microfluidic culture technology, both with single Organ Chips and fluidically coupled human "Body-on-Chips" platforms, which demonstrate their ability to recapitulate human physiology and disease states, as well as human patient responses to clinically relevant drug pharmacokinetic exposures, with higher fidelity than other in vitro models or animal studies. These findings raise the question of whether continuing to require results of animal testing for publication or grant funding still makes scientific or ethical sense, and if more physiologically relevant human Organ Chip models might better serve this purpose. This issue is addressed in this article in context of the history of the field, and advantages and disadvantages of Organ Chip approaches versus animal models are discussed that should be considered by the wider research community.
Collapse
Affiliation(s)
- Donald E. Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard UniversityBostonMA02115USA
- Vascular Biology Program, Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesCambridgeMA02138USA
| |
Collapse
|
209
|
Chen X, Zhang YS, Zhang X, Liu C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact Mater 2020; 6:1012-1027. [PMID: 33102943 PMCID: PMC7566214 DOI: 10.1016/j.bioactmat.2020.09.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine involves the use of engineered nanoscale materials in an extensive range of diagnostic and therapeutic applications and can be applied to the treatment of many diseases. Despite the rapid progress and tremendous potential of nanomedicine in the past decades, the clinical translational process is still quite slow, owing to the difficulty in understanding, evaluating, and predicting nanomaterial behaviors within the complex environment of human beings. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to resolve these challenges. Sophisticatedly designed Organ Chip enable in vitro simulation of the in vivo microenvironments, thus providing robust platforms for evaluating nanomedicine. Herein, we review recent developments and achievements in Organ Chip models for nanomedicine evaluations, categorized into seven broad sections based on the target organ systems: respiratory, digestive, lymphatic, excretory, nervous, and vascular, as well as coverage on applications relating to cancer. We conclude by providing our perspectives on the challenges and potential future directions for applications of Organ Chip in nanomedicine. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to understand, evaluate, and predict nanomedicine behaviors within the complex environment. Organ Chip models for nanomedicine evaluations are categorized into seven broad sections based on the targeted body systems. Limitations, challenges, and perspectives of Organ Chip for accelerating the assessment of nanomedicine are discussed, respectively.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, United States
| | - Xinping Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
210
|
Ashammakhi N, Nasiri R, Barros NRD, Tebon P, Thakor J, Goudie M, Shamloo A, Martin MG, Khademhosseini A. Gut-on-a-chip: Current progress and future opportunities. Biomaterials 2020; 255:120196. [PMID: 32623181 PMCID: PMC7396314 DOI: 10.1016/j.biomaterials.2020.120196] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/11/2020] [Accepted: 06/09/2020] [Indexed: 12/21/2022]
Abstract
Organ-on-a-chip technology tries to mimic the complexity of native tissues in vitro. Important progress has recently been made in using this technology to study the gut with and without microbiota. These in vitro models can serve as an alternative to animal models for studying physiology, pathology, and pharmacology. While these models have greater physiological relevance than two-dimensional (2D) cell systems in vitro, endocrine and immunological functions in gut-on-a-chip models are still poorly represented. Furthermore, the construction of complex models, in which different cell types and structures interact, remains a challenge. Generally, gut-on-a-chip models have the potential to advance our understanding of the basic interactions found within the gut and lay the foundation for future applications in understanding pathophysiology, developing drugs, and personalizing medical treatments.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Rohollah Nasiri
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Natan Roberto de Barros
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA.
| | - Peyton Tebon
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Jai Thakor
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Marcus Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Martin G Martin
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA, USA; Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California, Los Angeles, CA, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
211
|
Jeon B, Lee G, Wufuer M, Huang Y, Choi Y, Kim S, Choi TH. Enhanced predictive capacity using dual-parameter chip model that simulates physiological skin irritation. Toxicol In Vitro 2020; 68:104955. [DOI: 10.1016/j.tiv.2020.104955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/16/2020] [Accepted: 07/28/2020] [Indexed: 12/18/2022]
|
212
|
Aguilar-Rojas A, Olivo-Marin JC, Guillen N. Human intestinal models to study interactions between intestine and microbes. Open Biol 2020; 10:200199. [PMID: 33081633 PMCID: PMC7653360 DOI: 10.1098/rsob.200199] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Implementations of suitable in vitro cell culture systems of the human intestine have been essential tools in the study of the interaction among organs, commensal microbiota, pathogens and parasites. Due to the great complexity exhibited by the intestinal tissue, researchers have been developing in vitro/ex vivo systems to diminish the gap between conventional cell culture models and the human intestine. These models are able to reproduce different structures and functional aspects of the tissue. In the present review, information is recapitulated on the most used models, such as cell culture, intestinal organoids, scaffold-based three-dimensional models, and organ-on-a-chip and their use in studying the interaction between human intestine and microbes, and their advantages and limitations are also discussed.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Medicina Reproductiva, Unidad Médica de Alta Especialidad en Ginecología y Obstetricia No. 4 ‘Dr. Luis Castelazo Ayala’, Av. Río Magdalena No. 289, Col. Tizapán San Ángel, C.P. 01090 Ciudad de México, México
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, UMR3691, 25 Rue du Dr Roux, 75015 Paris, France
| | - Nancy Guillen
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, ERL9195, 25 Rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
213
|
Amigo L, Hernández-Ledesma B. Current Evidence on the Bioavailability of Food Bioactive Peptides. Molecules 2020; 25:E4479. [PMID: 33003506 PMCID: PMC7582556 DOI: 10.3390/molecules25194479] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 12/23/2022] Open
Abstract
Food protein-derived bioactive peptides are recognized as valuable ingredients of functional foods and/or nutraceuticals to promote health and reduce the risk of chronic diseases. However, although peptides have been demonstrated to exert multiple benefits by biochemical assays, cell culture, and animal models, the ability to translate the new findings into practical or commercial uses remains delayed. This fact is mainly due to the lack of correlation of in vitro findings with in vivo functions of peptides because of their low bioavailability. Once ingested, peptides need to resist the action of digestive enzymes during their transit through the gastrointestinal tract and cross the intestinal epithelial barrier to reach the target organs in an intact and active form to exert their health-promoting properties. Thus, for a better understanding of the in vivo physiological effects of food bioactive peptides, extensive research studies on their gastrointestinal stability and transport are needed. This review summarizes the most current evidence on those factors affecting the digestive and absorptive processes of food bioactive peptides, the recently designed models mimicking the gastrointestinal environment, as well as the novel strategies developed and currently applied to enhance the absorption and bioavailability of peptides.
Collapse
Affiliation(s)
| | - Blanca Hernández-Ledesma
- Department of Bioactivity and Food Analysis, Institute of Research in Food Sciences (CIAL, CSIC-UAM, CEI-UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain;
| |
Collapse
|
214
|
Kohl Y, Rundén-Pran E, Mariussen E, Hesler M, El Yamani N, Longhin EM, Dusinska M. Genotoxicity of Nanomaterials: Advanced In Vitro Models and High Throughput Methods for Human Hazard Assessment-A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1911. [PMID: 32992722 PMCID: PMC7601632 DOI: 10.3390/nano10101911] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
Changes in the genetic material can lead to serious human health defects, as mutations in somatic cells may cause cancer and can contribute to other chronic diseases. Genotoxic events can appear at both the DNA, chromosomal or (during mitosis) whole genome level. The study of mechanisms leading to genotoxicity is crucially important, as well as the detection of potentially genotoxic compounds. We consider the current state of the art and describe here the main endpoints applied in standard human in vitro models as well as new advanced 3D models that are closer to the in vivo situation. We performed a literature review of in vitro studies published from 2000-2020 (August) dedicated to the genotoxicity of nanomaterials (NMs) in new models. Methods suitable for detection of genotoxicity of NMs will be presented with a focus on advances in miniaturization, organ-on-a-chip and high throughput methods.
Collapse
Affiliation(s)
- Yvonne Kohl
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280 Sulzbach, Germany;
| | - Elise Rundén-Pran
- Health Effects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (E.M.); (N.E.Y.); (E.M.L.); (M.D.)
| | - Espen Mariussen
- Health Effects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (E.M.); (N.E.Y.); (E.M.L.); (M.D.)
| | - Michelle Hesler
- Fraunhofer Institute for Biomedical Engineering IBMT, 66280 Sulzbach, Germany;
| | - Naouale El Yamani
- Health Effects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (E.M.); (N.E.Y.); (E.M.L.); (M.D.)
| | - Eleonora Marta Longhin
- Health Effects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (E.M.); (N.E.Y.); (E.M.L.); (M.D.)
| | - Maria Dusinska
- Health Effects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway; (E.R.-P.); (E.M.); (N.E.Y.); (E.M.L.); (M.D.)
| |
Collapse
|
215
|
Morsink MAJ, Willemen NGA, Leijten J, Bansal R, Shin SR. Immune Organs and Immune Cells on a Chip: An Overview of Biomedical Applications. MICROMACHINES 2020; 11:mi11090849. [PMID: 32932680 PMCID: PMC7570325 DOI: 10.3390/mi11090849] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/27/2022]
Abstract
Understanding the immune system is of great importance for the development of drugs and the design of medical implants. Traditionally, two-dimensional static cultures have been used to investigate the immune system in vitro, while animal models have been used to study the immune system’s function and behavior in vivo. However, these conventional models do not fully emulate the complexity of the human immune system or the human in vivo microenvironment. Consequently, many promising preclinical findings have not been reproduced in human clinical trials. Organ-on-a-chip platforms can provide a solution to bridge this gap by offering human micro-(patho)physiological systems in which the immune system can be studied. This review provides an overview of the existing immune-organs-on-a-chip platforms, with a special emphasis on interorgan communication. In addition, future challenges to develop a comprehensive immune system-on-chip model are discussed.
Collapse
Affiliation(s)
- Margaretha A. J. Morsink
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Niels G. A. Willemen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell BioPhysics, Technical Medical Centre, Faculty of Science and Technology, University of Twente Drienerlolaan 5, 7522 NB Enschede, The Netherlands;
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA; (M.A.J.M.); (N.G.A.W.)
- Correspondence: ; Tel.: +1-617-768-8320
| |
Collapse
|
216
|
Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov 2020; 20:345-361. [PMID: 32913334 DOI: 10.1038/s41573-020-0079-3] [Citation(s) in RCA: 420] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Organs-on-chips (OoCs), also known as microphysiological systems or 'tissue chips' (the terms are synonymous), have attracted substantial interest in recent years owing to their potential to be informative at multiple stages of the drug discovery and development process. These innovative devices could provide insights into normal human organ function and disease pathophysiology, as well as more accurately predict the safety and efficacy of investigational drugs in humans. Therefore, they are likely to become useful additions to traditional preclinical cell culture methods and in vivo animal studies in the near term, and in some cases replacements for them in the longer term. In the past decade, the OoC field has seen dramatic advances in the sophistication of biology and engineering, in the demonstration of physiological relevance and in the range of applications. These advances have also revealed new challenges and opportunities, and expertise from multiple biomedical and engineering fields will be needed to fully realize the promise of OoCs for fundamental and translational applications. This Review provides a snapshot of this fast-evolving technology, discusses current applications and caveats for their implementation, and offers suggestions for directions in the next decade.
Collapse
Affiliation(s)
- Lucie A Low
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| | - Christine Mummery
- Leiden University Medical Center, Leiden, Netherlands.,University of Twente, Enschede, Netherlands
| | - Brian R Berridge
- National Institute for Environmental Health Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Christopher P Austin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Danilo A Tagle
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
217
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
218
|
Tan HY, Toh YC. What can microfluidics do for human microbiome research? BIOMICROFLUIDICS 2020; 14:051303. [PMID: 33062112 PMCID: PMC7538166 DOI: 10.1063/5.0012185] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/17/2020] [Indexed: 05/13/2023]
Abstract
Dysregulation of the human microbiome has been linked to various disease states, which has galvanized the efforts to modulate human health through microbiomes. Currently, human microbiome research is going through several phases to identify the constituent components of the microbiome, associate microbiome changes with physiological and pathological states, understand causative relationships, and finally translate this knowledge into therapeutics and diagnostics. The convergence of microfluidic technologies with molecular and cell profiling, microbiology, and tissue engineering can potentially be applied to these different phases of microbiome research to overcome the existing challenges faced by conventional approaches. The goal of this paper is to discuss and highlight the opportunities of applying different microfluidic technologies to specific areas of microbiome research as well as unique challenges that microfluidics must overcome when working with microbiome-relevant biological materials, e.g., micro-organisms, host tissues, and fluids. We will discuss the applicability of integrated microfluidic systems for processing biological samples for genomic sequencing analyses. For functional analysis of the microbiota, we will cover state-of-the-art microfluidic devices for microbiota cultivation and functional measurements. Finally, we highlight the use of organs-on-chips to model various microbiome-host tissue interactions. We envision that microfluidic technologies may hold great promise in advancing the knowledge on the interplay between microbiome and human health, as well as its eventual translation into microbiome-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hsih-Yin Tan
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 117599
| | - Yi-Chin Toh
- Author to whom correspondence should be addressed:
| |
Collapse
|
219
|
Roh TT, Chen Y, Rudolph S, Gee M, Kaplan DL. InVitro Models of Intestine Innate Immunity. Trends Biotechnol 2020; 39:274-285. [PMID: 32854949 DOI: 10.1016/j.tibtech.2020.07.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/18/2022]
Abstract
Animal models have delivered critical insights into mechanisms underlying the intestinal innate immune system; however, inherent differences exist between human and animal systems. To further understand the intestine innate immune system, there is a growing need for in vitro tissue model systems using human cells. A critical feature of in vitro cell and tissue models is the subepithelial environment, which contains additional cell types and includes 2D, microfluidic, organoid, and 3D tissue models. Where mouse models for the study of intestinal innate immune systems fall short, developments from in vitro models continue to grow in importance to aid efforts to understand this system in the context of disease and potential treatments.
Collapse
Affiliation(s)
- Terrence T Roh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Michelle Gee
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
220
|
3D In Vitro Human Organ Mimicry Devices for Drug Discovery, Development, and Assessment. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6187048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past few decades have shown significant advancement as complex in vitro humanized systems have substituted animal trials and 2D in vitro studies. 3D humanized platforms mimic the organs of interest with their stimulations (physical, electrical, chemical, and mechanical). Organ-on-chip devices, including in vitro modelling of 3D organoids, 3D microfabrication, and 3D bioprinted platforms, play an essential role in drug discovery, testing, and assessment. In this article, a thorough review is provided of the latest advancements in the area of organ-on-chip devices targeting liver, kidney, lung, gut, heart, skin, and brain mimicry devices for drug discovery, development, and/or assessment. The current strategies, fabrication methods, and the specific application of each device, as well as the advantages and disadvantages, are presented for each reported platform. This comprehensive review also provides some insights on the challenges and future perspectives for the further advancement of each organ-on-chip device.
Collapse
|
221
|
Shin W, Ambrosini YM, Shin YC, Wu A, Min S, Koh D, Park S, Kim S, Koh H, Kim HJ. Robust Formation of an Epithelial Layer of Human Intestinal Organoids in a Polydimethylsiloxane-Based Gut-on-a-Chip Microdevice. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2. [PMID: 33532747 PMCID: PMC7849371 DOI: 10.3389/fmedt.2020.00002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Polydimethylsiloxane (PDMS) is a silicone polymer that has been predominantly used in a human organ-on-a-chip microphysiological system. The hydrophobic surface of a microfluidic channel made of PDMS often results in poor adhesion of the extracellular matrix (ECM) as well as cell attachment. The surface modification by plasma or UV/ozone treatment in a PDMS-based device produces a hydrophilic surface that allows robust ECM coating and the reproducible attachment of human intestinal immortalized cell lines. However, these surface-activating methods have not been successful in forming a monolayer of the biopsy-derived primary organoid epithelium. Several existing protocols to grow human intestinal organoid cells in a PDMS microchannel are not always reproducibly operative due to the limited information. Here, we report an optimized methodology that enables robust and reproducible attachment of the intestinal organoid epithelium in a PDMS-based gut-on-a-chip. Among several reported protocols, we optimized a method by performing polyethyleneimine-based surface functionalization followed by the glutaraldehyde cross linking to activate the PDMS surface. Moreover, we discovered that the post-functionalization step contributes to provide uniform ECM deposition that allows to produce a robust attachment of the dissociated intestinal organoid epithelium in a PDMS-based microdevice. We envision that our optimized protocol may disseminate an enabling methodology to advance the integration of human organotypic cultures in a human organ-on-a-chip for patient-specific disease modeling.
Collapse
Affiliation(s)
- Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Yoko M Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Alexander Wu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Domin Koh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Sowon Park
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Kim
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hong Koh
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
222
|
Hewes SA, Wilson RL, Estes MK, Shroyer NF, Blutt SE, Grande-Allen KJ. In Vitro Models of the Small Intestine: Engineering Challenges and Engineering Solutions. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:313-326. [PMID: 32046599 PMCID: PMC7462033 DOI: 10.1089/ten.teb.2019.0334] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Pathologies affecting the small intestine contribute significantly to the disease burden of both the developing and the developed world, which has motivated investigation into the disease mechanisms through in vitro models. Although existing in vitro models recapitulate selected features of the intestine, various important aspects have often been isolated or omitted due to the anatomical and physiological complexity. The small intestine's intricate microanatomy, heterogeneous cell populations, steep oxygen gradients, microbiota, and intestinal wall contractions are often not included in in vitro experimental models of the small intestine, despite their importance in both intestinal biology and pathology. Known and unknown interdependencies between various physiological aspects necessitate more complex in vitro models. Microfluidic technology has made it possible to mimic the dynamic mechanical environment, signaling gradients, and other important aspects of small intestinal biology. This review presents an overview of the complexity of small intestinal anatomy and bioengineered models that recapitulate some of these physiological aspects.
Collapse
Affiliation(s)
- Sarah A. Hewes
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Reid L. Wilson
- Department of Bioengineering, Rice University, Houston, Texas, USA
- Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | |
Collapse
|
223
|
Sharma A, Raman V, Lee J, Forbes NS. Mucus blocks probiotics but increases penetration of motile pathogens and induces TNF-α and IL-8 secretion. Biotechnol Bioeng 2020; 117:2540-2555. [PMID: 32396232 PMCID: PMC7806204 DOI: 10.1002/bit.27383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/29/2022]
Abstract
The mucosal barrier in combination with innate immune system are the first line of defense against luminal bacteria at the intestinal mucosa. Dysfunction of the mucus layer and bacterial infiltration are linked to tissue inflammation and disease. To study host-bacterial interactions at the mucosal interface, we created an experimental model that contains luminal space, a mucus layer, an epithelial layer, and suspended immune cells. Reconstituted porcine small intestinal mucus formed an 880 ± 230 µm thick gel layer and had a porous structure. In the presence of mucus, sevenfold less probiotic and nonmotile VSL#3 bacteria transmigrated across the epithelial barrier compared to no mucus. The higher bacterial transmigration caused immune cell differentiation and increased the concentration of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNF-α; p < .01). Surprisingly, the mucus layer increased transmigration of pathogenic Salmonella and increased secretion of TNF-α and IL-8 (p < .05). Nonmotile, flagella knockout Salmonella had lower transmigration and caused lower IL-8 and TNF-α secretion (p < .05). These results demonstrate that motility enables pathogenic bacteria to cross the mucus and epithelial layers, which could lead to infection. Using an in vitro coculture platform to understand the interactions of bacteria with the intestinal mucosa has the potential to improve the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
224
|
Steinway SN, Saleh J, Koo BK, Delacour D, Kim DH. Human Microphysiological Models of Intestinal Tissue and Gut Microbiome. Front Bioeng Biotechnol 2020; 8:725. [PMID: 32850690 PMCID: PMC7411353 DOI: 10.3389/fbioe.2020.00725] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a complex system responsible for nutrient absorption, digestion, secretion, and elimination of waste products that also hosts immune surveillance, the intestinal microbiome, and interfaces with the nervous system. Traditional in vitro systems cannot harness the architectural and functional complexity of the GI tract. Recent advances in organoid engineering, microfluidic organs-on-a-chip technology, and microfabrication allows us to create better in vitro models of human organs/tissues. These micro-physiological systems could integrate the numerous cell types involved in GI development and physiology, including intestinal epithelium, endothelium (vascular), nerve cells, immune cells, and their interplay/cooperativity with the microbiome. In this review, we report recent progress in developing micro-physiological models of the GI systems. We also discuss how these models could be used to study normal intestinal physiology such as nutrient absorption, digestion, and secretion as well as GI infection, inflammation, cancer, and metabolism.
Collapse
Affiliation(s)
- Steven N. Steinway
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jad Saleh
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology, Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR 7592, Paris Diderot University, Paris, France
| | - Deok-Ho Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
225
|
Rapid Fabrication of Membrane-Integrated Thermoplastic Elastomer Microfluidic Devices. MICROMACHINES 2020; 11:mi11080731. [PMID: 32731570 PMCID: PMC7463978 DOI: 10.3390/mi11080731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/19/2020] [Accepted: 07/25/2020] [Indexed: 02/06/2023]
Abstract
Leveraging the advantageous material properties of recently developed soft thermoplastic elastomer materials, this work presents the facile and rapid fabrication of composite membrane-integrated microfluidic devices consisting of FlexdymTM polymer and commercially available porous polycarbonate membranes. The three-layer devices can be fabricated in under 2.5 h, consisting of a 2-min hot embossing cycle, conformal contact between device layers and a low-temperature baking step. The strength of the FlexdymTM-polycarbonate seal was characterized using a specialized microfluidic delamination device and an automated pressure controller configuration, offering a standardized and high-throughput method of microfluidic burst testing. Given a minimum bonding distance of 200 μm, the materials showed bonding that reliably withstood pressures of 500 mbar and above, which is sufficient for most microfluidic cell culture applications. Bonding was also stable when subjected to long term pressurization (10 h) and repeated use (10,000 pressure cycles). Cell culture trials confirmed good cell adhesion and sustained culture of human dermal fibroblasts on a polycarbonate membrane inside the device channels over the course of one week. In comparison to existing porous membrane-based microfluidic platforms of this configuration, most often made of polydimethylsiloxane (PDMS), these devices offer a streamlined fabrication methodology with materials having favourable properties for cell culture applications and the potential for implementation in barrier model organ-on-chips.
Collapse
|
226
|
Langerak N, Ahmed HMM, Li Y, Middel IR, Eslami Amirabadi H, Malda J, Masereeuw R, van Roij R. A Theoretical and Experimental Study to Optimize Cell Differentiation in a Novel Intestinal Chip. Front Bioeng Biotechnol 2020; 8:763. [PMID: 32793567 PMCID: PMC7393935 DOI: 10.3389/fbioe.2020.00763] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022] Open
Abstract
Microphysiological systems have potential as test systems in studying the intestinal barrier, in which shear stress is critical for the differentiation of Caco-2 cells into enterocytes. The most commonly used in vitro gut model for intestinal barrier studies is based on trans-well cultures. Albeit useful, these culture systems lack physiological shear stress which is believed to be critical for the differentiation of Caco-2 cells into enterocytes and to form tight monolayers. Conversely, organ-on-chip models have presented themselves as a promising alternative since it provides cells with the required shear stress. To this end, a novel biocompatible 3D-printed microfluidic device was developed. In this device, Caco-2 cells were seeded under physiologically-relevant unidirectional shear stress and compared to cells cultured under gravity-driven flow. Using numerical studies, the flow rate that corresponds to the required shear stress was calculated. Experimental tests were conducted to verify the effect of this on cell differentiation. The experiments clearly showed an enhancement of cell differentiation potential in a unidirectional physiologically-relevant pump-driven flow system (PDFS) as opposed to the simpler bidirectional gravity-driven flow system (GDFS). Additionally, computational modeling of an adapted design confirmed its ability to supply all cells with a more homogeneous shear stress, potentially further enhancing their differentiation. The shear stress in the adapted design can be well-approximated with analytic methods, thus allowing for efficient predictions for all parameter values in the system. The developed novel microfluidic device led to the formation of a tighter monolayer and enhanced functional properties of the differentiated Caco-2 cells, which presents a promising tool for preclinical in vitro testing of drugs in an animal-free platform.
Collapse
Affiliation(s)
- Nicky Langerak
- Institute for Theoretical Physics, Utrecht University, Utrecht, Netherlands
| | - Haysam M M Ahmed
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Yang Li
- Utrecht Biofabrication Facility, University Medical Center Utrecht, Utrecht, Netherlands
| | - Igor R Middel
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Hossein Eslami Amirabadi
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Human Cell Biology Group, Division of Metabolic Health Research, TNO, Zeist, Netherlands
| | - Jos Malda
- Utrecht Biofabrication Facility, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - René van Roij
- Institute for Theoretical Physics, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
227
|
Shin YC, Shin W, Koh D, Wu A, Ambrosini YM, Min S, Eckhardt SG, Fleming RYD, Kim S, Park S, Koh H, Yoo TK, Kim HJ. Three-Dimensional Regeneration of Patient-Derived Intestinal Organoid Epithelium in a Physiodynamic Mucosal Interface-on-a-Chip. MICROMACHINES 2020; 11:E663. [PMID: 32645991 PMCID: PMC7408321 DOI: 10.3390/mi11070663] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022]
Abstract
The regeneration of the mucosal interface of the human intestine is critical in the host-gut microbiome crosstalk associated with gastrointestinal diseases. The biopsy-derived intestinal organoids provide genetic information of patients with physiological cytodifferentiation. However, the enclosed lumen and static culture condition substantially limit the utility of patient-derived organoids for microbiome-associated disease modeling. Here, we report a patient-specific three-dimensional (3D) physiodynamic mucosal interface-on-a-chip (PMI Chip) that provides a microphysiological intestinal milieu under defined biomechanics. The real-time imaging and computational simulation of the PMI Chip verified the recapitulation of non-linear luminal and microvascular flow that simulates the hydrodynamics in a living human gut. The multiaxial deformations in a convoluted microchannel not only induced dynamic cell strains but also enhanced particle mixing in the lumen microchannel. Under this physiodynamic condition, an organoid-derived epithelium obtained from the patients diagnosed with Crohn's disease, ulcerative colitis, or colorectal cancer independently formed 3D epithelial layers with disease-specific differentiations. Moreover, co-culture with the human fecal microbiome in an anoxic-oxic interface resulted in the formation of stochastic microcolonies without a loss of epithelial barrier function. We envision that the patient-specific PMI Chip that conveys genetic, epigenetic, and environmental factors of individual patients will potentially demonstrate the pathophysiological dynamics and complex host-microbiome crosstalk to target a patient-specific disease modeling.
Collapse
Affiliation(s)
- Yong Cheol Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Domin Koh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Alexander Wu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Yoko M. Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
| | - S. Gail Eckhardt
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
| | - R. Y. Declan Fleming
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Seung Kim
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Sowon Park
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Hong Koh
- Severance Fecal Microbiota Transplantation Center, Severance Hospital, Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea; (S.K.); (S.P.); (H.K.)
| | - Tae Kyung Yoo
- Department of Computer Art, College of Art and Technology, Chung-Ang University, Seoul 06974, Korea;
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; (Y.C.S.); (W.S.); (D.K.); (A.W.); (Y.M.A.); (S.M.)
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; (S.G.E.); (R.Y.D.F.)
- Department of Medical Engineering, College of Medicine, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
228
|
Youhanna S, Lauschke VM. The Past, Present and Future of Intestinal In Vitro Cell Systems for Drug Absorption Studies. J Pharm Sci 2020; 110:50-65. [PMID: 32628951 DOI: 10.1016/j.xphs.2020.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/02/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022]
Abstract
The intestinal epithelium acts as a selective barrier for the absorption of water, nutrients and orally administered drugs. To evaluate the gastrointestinal permeability of a candidate molecule, scientists and drug developers have a multitude of cell culture models at their disposal. Static transwell cultures constitute the most extensively characterized intestinal in vitro system and can accurately categorize molecules into low, intermediate and high permeability compounds. However, they lack key aspects of intestinal physiology, including the cellular complexity of the intestinal epithelium, flow, mechanical strain, or interactions with intestinal mucus and microbes. To emulate these features, a variety of different culture paradigms, including microfluidic chips, organoids and intestinal slice cultures have been developed. Here, we provide an updated overview of intestinal in vitro cell culture systems and critically review their suitability for drug absorption studies. The available data show that these advanced culture models offer impressive possibilities for emulating intestinal complexity. However, there is a paucity of systematic absorption studies and benchmarking data and it remains unclear whether the increase in model complexity and costs translates into improved drug permeability predictions. In the absence of such data, conventional static transwell cultures remain the current gold-standard paradigm for drug absorption studies.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
229
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
230
|
Cassotta M, Forbes-Hernández TY, Calderón Iglesias R, Ruiz R, Elexpuru Zabaleta M, Giampieri F, Battino M. Links between Nutrition, Infectious Diseases, and Microbiota: Emerging Technologies and Opportunities for Human-Focused Research. Nutrients 2020; 12:E1827. [PMID: 32575399 PMCID: PMC7353391 DOI: 10.3390/nu12061827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
The interaction between nutrition and human infectious diseases has always been recognized. With the emergence of molecular tools and post-genomics, high-resolution sequencing technologies, the gut microbiota has been emerging as a key moderator in the complex interplay between nutrients, human body, and infections. Much of the host-microbial and nutrition research is currently based on animals or simplistic in vitro models. Although traditional in vivo and in vitro models have helped to develop mechanistic hypotheses and assess the causality of the host-microbiota interactions, they often fail to faithfully recapitulate the complexity of the human nutrient-microbiome axis in gastrointestinal homeostasis and infections. Over the last decade, remarkable progress in tissue engineering, stem cell biology, microfluidics, sequencing technologies, and computing power has taken place, which has produced a new generation of human-focused, relevant, and predictive tools. These tools, which include patient-derived organoids, organs-on-a-chip, computational analyses, and models, together with multi-omics readouts, represent novel and exciting equipment to advance the research into microbiota, infectious diseases, and nutrition from a human-biology-based perspective. After considering some limitations of the conventional in vivo and in vitro approaches, in this review, we present the main novel available and emerging tools that are suitable for designing human-oriented research.
Collapse
Affiliation(s)
- Manuela Cassotta
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Tamara Yuliett Forbes-Hernández
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
| | - Ruben Calderón Iglesias
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Roberto Ruiz
- Centre for Nutrition and Health, Universidad Europea del Atlántico (UEA), 39001 Santander, Spain; (M.C.); (R.C.I.); (R.R.)
| | - Maria Elexpuru Zabaleta
- Dipartimento di Scienze Cliniche e Molecolari, Facoltà di Medicina, Università Politecnica delle Marche, 60131 Ancona, Italy;
| | - Francesca Giampieri
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy
- College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Maurizio Battino
- Department of Analytical and Food Chemistry, Nutrition and Food Science Group, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
- Department of Clinical Sciences, Faculty of Medicine, Polytechnic University of Marche, 60131 Ancona, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
231
|
Azizipour N, Avazpour R, Rosenzweig DH, Sawan M, Ajji A. Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. MICROMACHINES 2020; 11:E599. [PMID: 32570945 PMCID: PMC7345732 DOI: 10.3390/mi11060599] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/21/2022]
Abstract
Following the advancements in microfluidics and lab-on-a-chip (LOC) technologies, a novel biomedical application for microfluidic based devices has emerged in recent years and microengineered cell culture platforms have been created. These micro-devices, known as organ-on-a-chip (OOC) platforms mimic the in vivo like microenvironment of living organs and offer more physiologically relevant in vitro models of human organs. Consequently, the concept of OOC has gained great attention from researchers in the field worldwide to offer powerful tools for biomedical researches including disease modeling, drug development, etc. This review highlights the background of biochip development. Herein, we focus on applications of LOC devices as a versatile tool for POC applications. We also review current progress in OOC platforms towards body-on-a-chip, and we provide concluding remarks and future perspectives for OOC platforms for POC applications.
Collapse
Affiliation(s)
- Neda Azizipour
- Institut de Génie Biomédical, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
| | - Rahi Avazpour
- Department of Chemical Engineering, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
| | - Derek H. Rosenzweig
- Department of Surgery, McGill University, Montreal, QC H3G 1A4, Canada;
- Injury, Repair and Recovery Program, Research Institute of McGill University Health Centre, Montreal, QC H3H 2R9, Canada
| | - Mohamad Sawan
- Polystim Neurotech Laboratory, Electrical Engineering Department, Polytechnique Montreal, QC H3T 1J4, Canada
- CenBRAIN Laboratory, School of Engineering, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, China
| | - Abdellah Ajji
- Institut de Génie Biomédical, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada;
- NSERC-Industry Chair, CREPEC, Chemical Engineering Department, Polytechnique Montreal, Montreal, QC H3C 3A7, Canada
| |
Collapse
|
232
|
Caruso G, Musso N, Grasso M, Costantino A, Lazzarino G, Tascedda F, Gulisano M, Lunte SM, Caraci F. Microfluidics as a Novel Tool for Biological and Toxicological Assays in Drug Discovery Processes: Focus on Microchip Electrophoresis. MICROMACHINES 2020; 11:E593. [PMID: 32549277 PMCID: PMC7344675 DOI: 10.3390/mi11060593] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
The last decades of biological, toxicological, and pharmacological research have deeply changed the way researchers select the most appropriate 'pre-clinical model'. The absence of relevant animal models for many human diseases, as well as the inaccurate prognosis coming from 'conventional' pre-clinical models, are among the major reasons of the failures observed in clinical trials. This evidence has pushed several research groups to move more often from a classic cellular or animal modeling approach to an alternative and broader vision that includes the involvement of microfluidic-based technologies. The use of microfluidic devices offers several benefits including fast analysis times, high sensitivity and reproducibility, the ability to quantitate multiple chemical species, and the simulation of cellular response mimicking the closest human in vivo milieu. Therefore, they represent a useful way to study drug-organ interactions and related safety and toxicity, and to model organ development and various pathologies 'in a dish'. The present review will address the applicability of microfluidic-based technologies in different systems (2D and 3D). We will focus our attention on applications of microchip electrophoresis (ME) to biological and toxicological studies as well as in drug discovery and development processes. These include high-throughput single-cell gene expression profiling, simultaneous determination of antioxidants and reactive oxygen and nitrogen species, DNA analysis, and sensitive determination of neurotransmitters in biological fluids. We will discuss new data obtained by ME coupled to laser-induced fluorescence (ME-LIF) and electrochemical detection (ME-EC) regarding the production and degradation of nitric oxide, a fundamental signaling molecule regulating virtually every critical cellular function. Finally, the integration of microfluidics with recent innovative technologies-such as organoids, organ-on-chip, and 3D printing-for the design of new in vitro experimental devices will be presented with a specific attention to drug development applications. This 'composite' review highlights the potential impact of 2D and 3D microfluidic systems as a fast, inexpensive, and highly sensitive tool for high-throughput drug screening and preclinical toxicological studies.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Margherita Grasso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Angelita Costantino
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Massimo Gulisano
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
- Molecular Preclinical and Translational Imaging Research Centre-IMPRonTE, University of Catania, 95125 Catania, Italy
- Interuniversity Consortium for Biotechnology, Area di Ricerca, Padriciano, 34149 Trieste, Italy
| | - Susan M. Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA;
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| |
Collapse
|
233
|
Wang K, Man K, Liu J, Liu Y, Chen Q, Zhou Y, Yang Y. Microphysiological Systems: Design, Fabrication, and Applications. ACS Biomater Sci Eng 2020; 6:3231-3257. [PMID: 33204830 PMCID: PMC7668566 DOI: 10.1021/acsbiomaterials.9b01667] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Microphysiological systems, including organoids, 3-D printed tissue constructs and organ-on-a-chips (organ chips), are physiologically relevant in vitro models and have experienced explosive growth in the past decades. Different from conventional, tissue culture plastic-based in vitro models or animal models, microphysiological systems recapitulate key microenvironmental characteristics of human organs and mimic their primary functions. The advent of microphysiological systems is attributed to evolving biomaterials, micro-/nanotechnologies and stem cell biology, which enable the precise control over the matrix properties and the interactions between cells, tissues and organs in physiological conditions. As such, microphysiological systems have been developed to model a broad spectrum of organs from microvasculature, eye, to lung and many others to understand human organ development and disease pathology and facilitate drug discovery. Multiorgans-on-a-chip systems have also been developed by integrating multiple associated organ chips in a single platform, which allows to study and employ the organ function in a systematic approach. Here we first discuss the design principles of microphysiological systems with a focus on the anatomy and physiology of organs, and then review the commonly used fabrication techniques and biomaterials for microphysiological systems. Subsequently, we discuss the recent development of microphysiological systems, and provide our perspectives on advancing microphysiological systems for preclinical investigation and drug discovery of human disease.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yang Liu
- North Texas Eye Research Institute, Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Qi Chen
- The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhou
- Department of Emergency, Xinqiao Hospital, Chongqing 400037, China
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
234
|
Wright L, Barnes TJ, Prestidge CA. Oral delivery of protein-based therapeutics: Gastroprotective strategies, physiological barriers and in vitro permeability prediction. Int J Pharm 2020; 585:119488. [PMID: 32504774 DOI: 10.1016/j.ijpharm.2020.119488] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
Abstract
The number of biological molecules emerging as therapeutics is growing exponentially due to their higher specificity and tolerability profiles compared to small molecules. Despite this, their traditionally parenteral delivery often results in poor patient compliance and incomplete treatment. Current research is focussed on developing effective oral delivery strategies to facilitate administration of these biomolecules, however no universal method exists to simultaneously provide gastric protection as well as enhance transport across the gastrointestinal epithelium. Furthermore, for efficient formulation development it is imperative that we can reliably analyse permeability of biomolecules through the gastrointestinal tract, highlighting the importance of the continual development and ongoing evaluation of in vitro predictive permeability tools. Here, we review the physiological obstacles associated with peptide and protein delivery throughout the gastrointestinal tract. Furthermore, we highlight methods utilised to circumvent these barriers and promote improved intestinal permeability. Lastly, we explore in vitro models employed to predict epithelial transport. Key findings highlight the need to carefully understand gastrointestinal physiology, allowing specific engineering of oral delivery systems for biomolecules. Significant importance is placed upon understanding enzymatic degradation susceptibility as well as uptake mechanisms for particulate and protein-based therapeutics for the development of successful oral protein delivery platforms.
Collapse
Affiliation(s)
- Leah Wright
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UniSA, Australia
| | - Timothy J Barnes
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia
| | - Clive A Prestidge
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UniSA, Australia.
| |
Collapse
|
235
|
Microfluidic chip for culturing intestinal epithelial cell layers: Characterization and comparison of drug transport between dynamic and static models. Toxicol In Vitro 2020; 65:104815. [DOI: 10.1016/j.tiv.2020.104815] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/28/2020] [Indexed: 12/29/2022]
|
236
|
Grau-Bové C, González-Quilen C, Terra X, Blay MT, Beltrán-Debón R, Jorba-Martín R, Espina B, Pinent M, Ardévol A. Effects of Flavanols on Enteroendocrine Secretion. Biomolecules 2020; 10:biom10060844. [PMID: 32492958 PMCID: PMC7355421 DOI: 10.3390/biom10060844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/23/2022] Open
Abstract
Some beneficial effects of grape seed proanthocyanidin extract (GSPE) can be explained by the modulation of enterohormone secretion. As GSPE comprises a combination of different molecules, the pure compounds that cause these effects need to be elucidated. The enterohormones and chemoreceptors present in the gastrointestinal tract differ between species, so if humans are to gain beneficial effects, species closer to humans-and humans themselves-must be used. We demonstrate that 100 mg/L of GSPE stimulates peptide YY (PYY) release, but not glucagon-like peptide 1 (GLP-1) release in the human colon. We used a pig ex vivo system that differentiates between apical and basolateral intestinal sides to analyse how apical stimulation with GSPE and its pure compounds affects the gastrointestinal tract. In pigs, apical GSPE treatment stimulates the basolateral release of PYY in the duodenum and colon and that of GLP-1 in the ascending, but not the descending colon. In the duodenum, luminal stimulation with procyanidin dimer B2 increased PYY secretion, but not CCK secretion, while catechin monomers (catechin/epicatechin) significantly increased CCK release, but not PYY release. The differential effects of GSPE and its pure compounds on enterohormone release at the same intestinal segment suggest that they act through chemosensors located apically and unevenly distributed along the gastrointestinal tract.
Collapse
Affiliation(s)
- Carme Grau-Bové
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
| | - Carlos González-Quilen
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
| | - Ximena Terra
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
| | - M. Teresa Blay
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
| | - Raul Beltrán-Debón
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
| | - Rosa Jorba-Martín
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
- Servei de Cirurgia General i de l’Aparell Digestiu, Hospital Universitari Joan XXIII, 43005 Tarragona, Spain
| | - Beatriz Espina
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
- Servei de Cirurgia General i de l’Aparell Digestiu, Hospital Universitari Joan XXIII, 43005 Tarragona, Spain
| | - Montserrat Pinent
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
- Correspondence: ; Tel.: +34-97-755-9566
| | - Anna Ardévol
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, c/Marcel·lí Domingo nº1, 43007 Tarragona, Spain; (C.G.-B.); (C.G.-Q.); (X.T.); (M.T.B.); (R.B.-D.); (A.A.)
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain; (R.J.-M.); (B.E.)
| |
Collapse
|
237
|
Hawkins KG, Casolaro C, Brown JA, Edwards DA, Wikswo JP. The Microbiome and the Gut-Liver-Brain Axis for Central Nervous System Clinical Pharmacology: Challenges in Specifying and Integrating In Vitro and In Silico Models. Clin Pharmacol Ther 2020; 108:929-948. [PMID: 32347548 PMCID: PMC7572575 DOI: 10.1002/cpt.1870] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
The complexity of integrating microbiota into clinical pharmacology, environmental toxicology, and opioid studies arises from bidirectional and multiscale interactions between humans and their many microbiota, notably those of the gut. Hosts and each microbiota are governed by distinct central dogmas, with genetics influencing transcriptomics, proteomics, and metabolomics. Each microbiota's metabolome differentially modulates its own and the host's multi‐omics. Exogenous compounds (e.g., drugs and toxins), often affect host multi‐omics differently than microbiota multi‐omics, shifting the balance between drug efficacy and toxicity. The complexity of the host‐microbiota connection has been informed by current methods of in vitro bacterial cultures and in vivo mouse models, but they fail to elucidate mechanistic details. Together, in vitro organ‐on‐chip microphysiological models, multi‐omics, and in silico computational models have the potential to supplement the established methods to help clinical pharmacologists and environmental toxicologists unravel the myriad of connections between the gut microbiota and host health and disease.
Collapse
Affiliation(s)
- Kyle G Hawkins
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA
| | - Caleb Casolaro
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jacquelyn A Brown
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - David A Edwards
- Department of Anesthesiology and Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John P Wikswo
- Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
238
|
Guo Y, Deng P, Chen W, Li Z. Modeling Pharmacokinetic Profiles for Assessment of Anti-Cancer Drug on a Microfluidic System. MICROMACHINES 2020; 11:E551. [PMID: 32486116 PMCID: PMC7344513 DOI: 10.3390/mi11060551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022]
Abstract
The pharmacokinetic (PK) properties of drug, which include drug absorption and excretion, play an important role in determining the in vivo pharmaceutical activity. However, current in vitro systems that model PK profiles are often limited by the in vivo-like concentration profile of a drug. Herein, we present a perfused and multi-layered microfluidic chip system to model the PK profile of anti-cancer drug 5-FU in vitro. The chip device contains two layers of culture channels sandwiched by a porous membrane, which allows for drug exposure and diffusion between the two channels. The integration of upper intestine cells (Caco-2) and bottom targeted cells within the device enables the generation of loading and clearance portions of a PK curve under peristaltic flow. Fluorescein as a test molecule was initially used to generate a concentration-time curve, investigating the effects of parameters of flow rate, administration time, and initial concentration on dynamic drug concentration profiles. Furthermore, anti-cancer drug 5-FU was performed to assess its pharmaceutical activity on target cells (human lung adenocarcinoma cells or human pulmonary alveolar epithelial cells) using different drug administration regimens. A dynamic, in vivo-like 5-FU exposure refers to PK profile regimen, led to generate a lower drug concentration (dynamically fluctuate from 0 to 1 μg/mL affected by absorption) compared to the constant exposure. Moreover, the PK profile regimen alleviates the drug-induced cytotoxicity on target cells. These results demonstrate the feasibility of determining the PK profiles using this microfluidic system with in vivo-like drug administration regimens. This established system may provide a powerful platform for the prediction of drug safety and effectiveness in the pharmaceutical research.
Collapse
Affiliation(s)
- Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.G.); (P.D.); (W.C.)
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Pengwei Deng
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.G.); (P.D.); (W.C.)
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Wenwen Chen
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.G.); (P.D.); (W.C.)
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Zhongyu Li
- Division of Biotechnology, CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China; (Y.G.); (P.D.); (W.C.)
- College of Life Science, Dalian Minzu University, Dalian 116600, China
| |
Collapse
|
239
|
Griffith CM, Huang SA, Cho C, Khare TM, Rich M, Lee GH, Ligler FS, Diekman BO, Polacheck WJ. Microfluidics for the study of mechanotransduction. JOURNAL OF PHYSICS D: APPLIED PHYSICS 2020; 53:224004. [PMID: 33840837 PMCID: PMC8034607 DOI: 10.1088/1361-6463/ab78d4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Mechanical forces regulate a diverse set of biological processes at cellular, tissue, and organismal length scales. Investigating the cellular and molecular mechanisms that underlie the conversion of mechanical forces to biological responses is challenged by limitations of traditional animal models and in vitro cell culture, including poor control over applied force and highly artificial cell culture environments. Recent advances in fabrication methods and material processing have enabled the development of microfluidic platforms that provide precise control over the mechanical microenvironment of cultured cells. These devices and systems have proven to be powerful for uncovering and defining mechanisms of mechanotransduction. In this review, we first give an overview of the main mechanotransduction pathways that function at sites of cell adhesion, many of which have been investigated with microfluidics. We then discuss how distinct microfluidic fabrication methods can be harnessed to gain biological insight, with description of both monolithic and replica molding approaches. Finally, we present examples of how microfluidics can be used to apply both solid forces (substrate mechanics, strain, and compression) and fluid forces (luminal, interstitial) to cells. Throughout the review, we emphasize the advantages and disadvantages of different fabrication methods and applications of force in order to provide perspective to investigators looking to apply forces to cells in their own research.
Collapse
Affiliation(s)
- Christian M Griffith
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Stephanie A Huang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Crescentia Cho
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Tanmay M Khare
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC
| | - Matthew Rich
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Gi-Hun Lee
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Frances S Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
| | - Brian O Diekman
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC
- McAllister Heart Institute, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
- Cancer Cell Biology Program, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| |
Collapse
|
240
|
Shanti A, Samara B, Abdullah A, Hallfors N, Accoto D, Sapudom J, Alatoom A, Teo J, Danti S, Stefanini C. Multi-Compartment 3D-Cultured Organ-on-a-Chip: Towards a Biomimetic Lymph Node for Drug Development. Pharmaceutics 2020; 12:E464. [PMID: 32438634 PMCID: PMC7284904 DOI: 10.3390/pharmaceutics12050464] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 12/23/2022] Open
Abstract
The interaction of immune cells with drugs and/or with other cell types should be mechanistically investigated in order to reduce attrition of new drug development. However, they are currently only limited technologies that address this need. In our work, we developed initial but significant building blocks that enable such immune-drug studies. We developed a novel microfluidic platform replicating the Lymph Node (LN) microenvironment called LN-on-a-chip, starting from design all the way to microfabrication, characterization and validation in terms of architectural features, fluidics, cytocompatibility, and usability. To prove the biomimetics of this microenvironment, we inserted different immune cell types in a microfluidic device, which showed an in-vivo-like spatial distribution. We demonstrated that the developed LN-on-a-chip incorporates key features of the native human LN, namely, (i) similarity in extracellular matrix composition, morphology, porosity, stiffness, and permeability, (ii) compartmentalization of immune cells within distinct structural domains, (iii) replication of the lymphatic fluid flow pattern, (iv) viability of encapsulated cells in collagen over the typical timeframe of immunotoxicity experiments, and (v) interaction among different cell types across chamber boundaries. Further studies with this platform may assess the immune cell function as a step forward to disclose the effects of pharmaceutics to downstream immunology in more physiologically relevant microenvironments.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, UAE; (A.S.); (B.S.); (A.A.); (N.H.)
| | - Bisan Samara
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, UAE; (A.S.); (B.S.); (A.A.); (N.H.)
| | - Amal Abdullah
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, UAE; (A.S.); (B.S.); (A.A.); (N.H.)
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, UAE; (A.S.); (B.S.); (A.A.); (N.H.)
| | - Dino Accoto
- School of Mechanical & Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore;
| | - Jiranuwat Sapudom
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE; (J.S.); (A.A.); (J.T.)
| | - Aseel Alatoom
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE; (J.S.); (A.A.); (J.T.)
| | - Jeremy Teo
- Division of Engineering, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, UAE; (J.S.); (A.A.); (J.T.)
- Department of Biomedical and Mechanical Engineering, New York University, P.O. Box 903, New York, NY 10276-0903, USA
| | - Serena Danti
- Department of Civil and Industrial Engineering, University of Pisa, 56122 Pisa, Italy;
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, UAE; (A.S.); (B.S.); (A.A.); (N.H.)
| |
Collapse
|
241
|
Wodzanowski KA, Cassel SE, Grimes CL, Kloxin AM. Tools for probing host-bacteria interactions in the gut microenvironment: From molecular to cellular levels. Bioorg Med Chem Lett 2020; 30:127116. [PMID: 32223923 PMCID: PMC7476074 DOI: 10.1016/j.bmcl.2020.127116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/28/2020] [Accepted: 03/15/2020] [Indexed: 12/31/2022]
Abstract
Healthy function of the gut microenvironment is dependent on complex interactions between the bacteria of the microbiome, epithelial and immune (host) cells, and the surrounding tissue. Misregulation of these interactions is implicated in disease. A range of tools have been developed to study these interactions, from mechanistic studies to therapeutic evaluation. In this Digest, we highlight select tools at the cellular and molecular level for probing specific cell-microenvironment interactions. Approaches are overviewed for controlling and probing cell-cell interactions, from transwell and microfluidic devices to engineered bacterial peptidoglycan fragments, and cell-matrix interactions, from three-dimensional scaffolds to chemical handles for in situ modifications.
Collapse
Affiliation(s)
| | - Samantha E Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Catherine L Grimes
- Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, United States; Biological Sciences, University of Delaware, Newark, DE 19716, United States.
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States; Materials Science and Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
242
|
Mandsberg NK, Christfort JF, Kamguyan K, Boisen A, Srivastava SK. Orally ingestible medical devices for gut engineering. Adv Drug Deliv Rev 2020; 165-166:142-154. [PMID: 32416112 PMCID: PMC7255201 DOI: 10.1016/j.addr.2020.05.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/01/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022]
Abstract
Orally ingestible medical devices provide significant advancement for diagnosis and treatment of gastrointestinal (GI) tract-related conditions. From micro- to macroscale devices, with designs ranging from very simple to complex, these medical devices can be used for site-directed drug delivery in the GI tract, real-time imaging and sensing of gut biomarkers. Equipped with uni-direction release, or self-propulsion, or origami design, these microdevices are breaking the barriers associated with drug delivery, including biologics, across the GI tract. Further, on-board microelectronics allow imaging and sensing of gut tissue and biomarkers, providing a more comprehensive understanding of underlying pathophysiological conditions. We provide an overview of recent advances in orally ingestible medical devices towards drug delivery, imaging and sensing. Challenges associated with gut microenvironment, together with various activation/actuation modalities of medical devices for micromanipulation of the gut are discussed. We have critically examined the relationship between materials–device design–pharmacological responses with respect to existing regulatory guidelines and provided a clear roadmap for the future.
Collapse
|
243
|
Gunasekera S, Zahedi A, O’Dea M, King B, Monis P, Thierry B, M. Carr J, Ryan U. Organoids and Bioengineered Intestinal Models: Potential Solutions to the Cryptosporidium Culturing Dilemma. Microorganisms 2020; 8:microorganisms8050715. [PMID: 32403447 PMCID: PMC7285185 DOI: 10.3390/microorganisms8050715] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Cryptosporidium is a major cause of severe diarrhea-related disease in children in developing countries, but currently no vaccine or effective treatment exists for those who are most at risk of serious illness. This is partly due to the lack of in vitro culturing methods that are able to support the entire Cryptosporidium life cycle, which has led to research in Cryptosporidium biology lagging behind other protozoan parasites. In vivo models such as gnotobiotic piglets are complex, and standard in vitro culturing methods in transformed cell lines, such as HCT-8 cells, have not been able to fully support fertilization occurring in vitro. Additionally, the Cryptosporidium life cycle has also been reported to occur in the absence of host cells. Recently developed bioengineered intestinal models, however, have shown more promising results and are able to reproduce a whole cycle of infectivity in one model system. This review evaluates the recent advances in Cryptosporidium culturing techniques and proposes future directions for research that may build upon these successes.
Collapse
Affiliation(s)
- Samantha Gunasekera
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
- Correspondence: (S.G.); (U.R.); Tel.: +61-8-9360-2495 (S.G.); +61-8-9360-2482 (U.R.)
| | - Alireza Zahedi
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
| | - Mark O’Dea
- Antimicrobial Resistance and Infectious Diseases Laboratory, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia; m.o’
| | - Brendon King
- South Australian Water Corporation, Adelaide 5000, South Australia, Australia; (B.K.); (P.M.)
- College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia;
| | - Paul Monis
- South Australian Water Corporation, Adelaide 5000, South Australia, Australia; (B.K.); (P.M.)
- Future Industries Institute and ARC Centre of Excellence for Convergent Bio and Nano Science, University of South Australia, Adelaide 5095, South Australia, Australia;
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence for Convergent Bio and Nano Science, University of South Australia, Adelaide 5095, South Australia, Australia;
| | - Jillian M. Carr
- College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia;
| | - Una Ryan
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
- Correspondence: (S.G.); (U.R.); Tel.: +61-8-9360-2495 (S.G.); +61-8-9360-2482 (U.R.)
| |
Collapse
|
244
|
Shin W, Hackley LA, Kim HJ. "Good Fences Make Good Neighbors": How does the Human Gut Microchip Unravel Mechanism of Intestinal Inflammation? Gut Microbes 2020; 11:581-586. [PMID: 31198078 PMCID: PMC7524309 DOI: 10.1080/19490976.2019.1626684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A microengineered human gut-on-a-chip has demonstrated intestinal physiology, three-dimensional (3D) epithelial morphogenesis, and longitudinal host-microbiome interactions in vitro. The modular accessibility and modularity of the microphysiological gut-on-a-chip can lead to the identification of the seminal trigger in intestinal inflammation. By coupling microbial and immune cells in a spatiotemporal manner, we discovered that the maintenance of healthy epithelial barrier function is necessary and sufficient to demonstrate the homeostatic tolerance of the gut. Here, we highlight the breakthrough of our new disease model and discuss the future impact of investigating the etiology and therapeutic targets in the multifactorial inflammatory bowel disease.
Collapse
Affiliation(s)
- Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Landon A. Hackley
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA,CONTACT Hyun Jung Kim Department of Biomedical Engineering, The University of Texas at Austin, 107 W. Dean Keeton St., BME 4.202C, Austin, TX78712, USA
| |
Collapse
|
245
|
Ambrosini YM, Park Y, Jergens AE, Shin W, Min S, Atherly T, Borcherding DC, Jang J, Allenspach K, Mochel JP, Kim HJ. Recapitulation of the accessible interface of biopsy-derived canine intestinal organoids to study epithelial-luminal interactions. PLoS One 2020; 15:e0231423. [PMID: 32302323 PMCID: PMC7164685 DOI: 10.1371/journal.pone.0231423] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Recent advances in canine intestinal organoids have expanded the option for building a better in vitro model to investigate translational science of intestinal physiology and pathology between humans and animals. However, the three-dimensional geometry and the enclosed lumen of canine intestinal organoids considerably hinder the access to the apical side of epithelium for investigating the nutrient and drug absorption, host-microbiome crosstalk, and pharmaceutical toxicity testing. Thus, the creation of a polarized epithelial interface accessible from apical or basolateral side is critical. Here, we demonstrated the generation of an intestinal epithelial monolayer using canine biopsy-derived colonic organoids (colonoids). We optimized the culture condition to form an intact monolayer of the canine colonic epithelium on a nanoporous membrane insert using the canine colonoids over 14 days. Transmission and scanning electron microscopy revealed a physiological brush border interface covered by the microvilli with glycocalyx, as well as the presence of mucin granules, tight junctions, and desmosomes. The population of stem cells as well as differentiated lineage-dependent epithelial cells were verified by immunofluorescence staining and RNA in situ hybridization. The polarized expression of P-glycoprotein efflux pump was confirmed at the apical membrane. Also, the epithelial monolayer formed tight- and adherence-junctional barrier within 4 days, where the transepithelial electrical resistance and apparent permeability were inversely correlated. Hence, we verified the stable creation, maintenance, differentiation, and physiological function of a canine intestinal epithelial barrier, which can be useful for pharmaceutical and biomedical researches.
Collapse
Affiliation(s)
- Yoko M. Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States of America
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States of America
| | - Yejin Park
- Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Korea
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States of America
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States of America
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States of America
| | - Todd Atherly
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States of America
| | - Dana C. Borcherding
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States of America
| | - Jinah Jang
- Department of Creative IT Engineering, Pohang University of Science and Technology, Pohang, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Korea
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States of America
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States of America
- * E-mail: (HJK); (JPM)
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States of America
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, United States of America
- * E-mail: (HJK); (JPM)
| |
Collapse
|
246
|
Ambrosini YM, Shin W, Min S, Kim HJ. Microphysiological Engineering of Immune Responses in Intestinal Inflammation. Immune Netw 2020; 20:e13. [PMID: 32395365 PMCID: PMC7192834 DOI: 10.4110/in.2020.20.e13] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/18/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
The epithelial barrier in the gastrointestinal (GI) tract is a protective interface that endures constant exposure to the external environment while maintaining its close contact with the local immune system. Growing evidence has suggested that the intercellular crosstalk in the GI tract contributes to maintaining the homeostasis in coordination with the intestinal microbiome as well as the tissue-specific local immune elements. Thus, it is critical to map the complex crosstalks in the intestinal epithelial-microbiome-immune (EMI) axis to identify a pathological trigger in the development of intestinal inflammation, including inflammatory bowel disease. However, deciphering a specific contributor to the onset of pathophysiological cascades has been considerably hindered by the challenges in current in vivo and in vitro models. Here, we introduce various microphysiological engineering models of human immune responses in the EMI axis under the healthy conditions and gut inflammation. As a prospective model, we highlight how the human “gut inflammation-on-a-chip” can reconstitute the pathophysiological immune responses and contribute to understanding the independent role of inflammatory factors in the EMI axis on the initiation of immune responses under barrier dysfunction. We envision that the microengineered immune models can be useful to build a customizable patient's chip for the advance in precision medicine.
Collapse
Affiliation(s)
- Yoko M Ambrosini
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Woojung Shin
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Soyoun Min
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Hyun Jung Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
247
|
Winkler TE, Feil M, Stronkman EFGJ, Matthiesen I, Herland A. Low-cost microphysiological systems: feasibility study of a tape-based barrier-on-chip for small intestine modeling. LAB ON A CHIP 2020; 20:1212-1226. [PMID: 32141461 DOI: 10.1039/d0lc00009d] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We see affordability as a key challenge in making organs-on-chips accessible to a wider range of users, particularly outside the highest-resource environments. Here, we present an approach to barrier-on-a-chip fabrication based on double-sided pressure-sensitive adhesive tape and off-the-shelf polycarbonate. Besides a low materials cost, common also to PDMS or thermoplastics, it requires minimal (€100) investment in laboratory equipment, yet at the same time is suitable for upscaling to industrial roll-to-roll manufacture. We evaluate our microphysiological system with an epithelial (Caco-2/BBe1) barrier model of the small intestine, studying the biological effects of permeable support pore size, as well as stimulation with a common food compound (chili pepper-derived capsaicinoids). The cells form tight and continuous barrier layers inside our systems, with comparable permeability but superior epithelial polarization compared to Transwell culture, in line with other perfused microphysiological models. Permeable support pore size is shown to weakly impact barrier layer integrity as well as the metabolic cell profile. Capsaicinoid response proves distinct between culture systems, but we show that impacted metabolic pathways are partly conserved, and that cytoskeletal changes align with previous studies. Overall, our tape-based microphysiological system proves to be a robust and reproducible approach to studying physiological barriers, in spite of its low cost.
Collapse
Affiliation(s)
- Thomas E Winkler
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
248
|
Peters MF, Choy AL, Pin C, Leishman DJ, Moisan A, Ewart L, Guzzie-Peck PJ, Sura R, Keller DA, Scott CW, Kolaja KL. Developing in vitro assays to transform gastrointestinal safety assessment: potential for microphysiological systems. LAB ON A CHIP 2020; 20:1177-1190. [PMID: 32129356 DOI: 10.1039/c9lc01107b] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Drug-induced gastrointestinal toxicities (DI-GITs) are among the most common adverse events in clinical trials. High prevalence of DI-GIT has persisted among new drugs due in part to the lack of robust experimental tools to allow early detection or to guide optimization of safer molecules. Developing in vitro assays for the leading GI toxicities (nausea, vomiting, diarrhoea, constipation, and abdominal pain) will likely involve recapitulating complex physiological properties that require contributions from diverse cell/tissue types including epithelial, immune, microbiome, nerve, and muscle. While this stipulation may be beyond traditional 2D monocultures of intestinal cell lines, emerging 3D GI microtissues capture interactions between diverse cell and tissue types. These interactions give rise to microphysiologies fundamental to gut biology. For GI microtissues, organoid technology was the breakthrough that introduced intestinal stem cells with the capability of differentiating into each of the epithelial cell types and that self-organize into a multi-cellular tissue proxy with villus- and crypt-like domains. Recently, GI microtissues generated using miniaturized devices with microfluidic flow and cyclic peristaltic strain were shown to induce Caco2 cells to spontaneously differentiate into each of the principle intestinal epithelial cell types. Second generation models comprised of epithelial organoids or microtissues co-cultured with non-epithelial cell types can successfully reproduce cross-'tissue' functional interactions broadening the potential of these models to accurately study drug-induced toxicities. A new paradigm in which in vitro assays become an early part of GI safety assessment could be realized if microphysiological systems (MPS) are developed in alignment with drug-discovery needs. Herein, approaches for assessing GI toxicity of pharmaceuticals are reviewed and gaps are compared with capabilities of emerging GI microtissues (e.g., organoids, organ-on-a-chip, transwell systems) in order to provide perspective on the assay features needed for MPS models to be adopted for DI-GIT assessment.
Collapse
Affiliation(s)
- Matthew F Peters
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Boston, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Cong Y, Han X, Wang Y, Chen Z, Lu Y, Liu T, Wu Z, Jin Y, Luo Y, Zhang X. Drug Toxicity Evaluation Based on Organ-on-a-chip Technology: A Review. MICROMACHINES 2020; 11:E381. [PMID: 32260191 PMCID: PMC7230535 DOI: 10.3390/mi11040381] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
Organ-on-a-chip academic research is in its blossom. Drug toxicity evaluation is a promising area in which organ-on-a-chip technology can apply. A unique advantage of organ-on-a-chip is the ability to integrate drug metabolism and drug toxic processes in a single device, which facilitates evaluation of toxicity of drug metabolites. Human organ-on-a-chip has been fabricated and used to assess drug toxicity with data correlation with the clinical trial. In this review, we introduced the microfluidic chip models of liver, kidney, heart, nerve, and other organs and multiple organs, highlighting the application of these models in drug toxicity detection. Some biomarkers of toxic injury that have been used in organ chip platforms or have potential for use on organ chip platforms are summarized. Finally, we discussed the goals and future directions for drug toxicity evaluation based on organ-on-a-chip technology.
Collapse
Affiliation(s)
- Ye Cong
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116023, China;
| | - Xiahe Han
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| | - Youping Wang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| | - Zongzheng Chen
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yao Lu
- Biotechnologhy Division, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian 116011, China;
| | - Zhengzhi Wu
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yu Jin
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116023, China;
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| |
Collapse
|
250
|
Volpe DA. Advances in cell-based permeability assays to screen drugs for intestinal absorption. Expert Opin Drug Discov 2020; 15:539-549. [DOI: 10.1080/17460441.2020.1735347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Donna A. Volpe
- Division of Applied Regulatory Science, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|