51
|
Namdee K, Khongkow M, Boonthod S, Boonrungsiman S, Jarussophon S, Pongwan P, Yata T, Saengkrit N. Cell-based assay for characterizing cell adhesion properties of active targeted nanoparticles under static and flow condition using an integrated flow chamber. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
52
|
Watson DE, Hunziker R, Wikswo JP. Fitting tissue chips and microphysiological systems into the grand scheme of medicine, biology, pharmacology, and toxicology. Exp Biol Med (Maywood) 2017; 242:1559-1572. [PMID: 29065799 DOI: 10.1177/1535370217732765] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Microphysiological systems (MPS), which include engineered organoids (EOs), single organ/tissue chips (TCs), and multiple organs interconnected to create miniature in vitro models of human physiological systems, are rapidly becoming effective tools for drug development and the mechanistic understanding of tissue physiology and pathophysiology. The second MPS thematic issue of Experimental Biology and Medicine comprises 15 articles by scientists and engineers from the National Institutes of Health, the IQ Consortium, the Food and Drug Administration, and Environmental Protection Agency, an MPS company, and academia. Topics include the progress, challenges, and future of organs-on-chips, dissemination of TCs into Pharma, children's health protection, liver zonation, liver chips and their coupling to interconnected systems, gastrointestinal MPS, maturation of immature cardiomyocytes in a heart-on-a-chip, coculture of multiple cell types in a human skin construct, use of synthetic hydrogels to create EOs that form neural tissue models, the blood-brain barrier-on-a-chip, MPS models of coupled female reproductive organs, coupling MPS devices to create a body-on-a-chip, and the use of a microformulator to recapitulate endocrine circadian rhythms. While MPS hardware has been relatively stable since the last MPS thematic issue, there have been significant advances in cell sourcing, with increased reliance on human-induced pluripotent stem cells, and in characterization of the genetic and functional cell state in MPS bioreactors. There is growing appreciation of the need to minimize perfusate-to-cell-volume ratios and respect physiological scaling of coupled TCs. Questions asked by drug developers are followed by an analysis of the potential value, costs, and needs of Pharma. Of highest value and lowest switching costs may be the development of MPS disease models to aid in the discovery of disease mechanisms; novel compounds including probes, leads, and clinical candidates; and mechanism of action of drug candidates. Impact statement Microphysiological systems (MPS), which include engineered organoids and both individual and coupled organs-on-chips and tissue chips, are a rapidly growing topic of research that addresses the known limitations of conventional cellular monoculture on flat plastic - a well-perfected set of techniques that produces reliable, statistically significant results that may not adequately represent human biology and disease. As reviewed in this article and the others in this thematic issue, MPS research has made notable progress in the past three years in both cell sourcing and characterization. As the field matures, currently identified challenges are being addressed, and new ones are being recognized. Building upon investments by the Defense Advanced Research Projects Agency, National Institutes of Health, Food and Drug Administration, Defense Threat Reduction Agency, and Environmental Protection Agency of more than $200 million since 2012 and sizable corporate spending, academic and commercial players in the MPS community are demonstrating their ability to meet the translational challenges required to apply MPS technologies to accelerate drug development and advance toxicology.
Collapse
Affiliation(s)
| | - Rosemarie Hunziker
- 2 National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - John P Wikswo
- 3 Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235-1807, USA
| |
Collapse
|
53
|
Costello CM, Phillipsen MB, Hartmanis LM, Kwasnica MA, Chen V, Hackam D, Chang MW, Bentley WE, March JC. Microscale Bioreactors for in situ characterization of GI epithelial cell physiology. Sci Rep 2017; 7:12515. [PMID: 28970586 PMCID: PMC5624909 DOI: 10.1038/s41598-017-12984-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 09/14/2017] [Indexed: 02/08/2023] Open
Abstract
The development of in vitro artificial small intestines that realistically mimic in vivo systems will enable vast improvement of our understanding of the human gut and its impact on human health. Synthetic in vitro models can control specific parameters, including (but not limited to) cell types, fluid flow, nutrient profiles and gaseous exchange. They are also “open” systems, enabling access to chemical and physiological information. In this work, we demonstrate the importance of gut surface topography and fluid flow dynamics which are shown to impact epithelial cell growth, proliferation and intestinal cell function. We have constructed a small intestinal bioreactor using 3-D printing and polymeric scaffolds that mimic the 3-D topography of the intestine and its fluid flow. Our results indicate that TEER measurements, which are typically high in static 2-D Transwell apparatuses, is lower in the presence of liquid sheer and 3-D topography compared to a flat scaffold and static conditions. There was also increased cell proliferation and discovered localized regions of elevated apoptosis, specifically at the tips of the villi, where there is highest sheer. Similarly, glucose was actively transported (as opposed to passive) and at higher rates under flow.
Collapse
Affiliation(s)
- Cait M Costello
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA
| | - Mikkel B Phillipsen
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA
| | - Leonard M Hartmanis
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA
| | - Marek A Kwasnica
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA
| | - Victor Chen
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA
| | - David Hackam
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, USA
| | - Matthew W Chang
- Department of Biochemistry, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - William E Bentley
- Institute for Biomedical Devices, University of Maryland, Maryland, USA
| | - John C March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, USA.
| |
Collapse
|
54
|
Wang Y, Gunasekara DB, Attayek PJ, Reed MI, DiSalvo M, Nguyen DL, Dutton JS, Lebhar MS, Bultman SJ, Sims CE, Magness ST, Allbritton NL. In Vitro Generation of Mouse Colon Crypts. ACS Biomater Sci Eng 2017; 3:2502-2513. [PMID: 30854421 DOI: 10.1021/acsbiomaterials.7b00368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Organoid culture has had a significant impact on in vitro studies of the intestinal epithelium; however, the exquisite architecture, luminal accessibility, and lineage compartmentalization found in vivo has not been recapitulated in the organoid systems. We have used a microengineered platform with suitable extracellular matrix contacts and stiffness to generate a self-renewing mouse colonic epithelium that replicates key architectural and physiological functions found in vivo, including a surface lined with polarized crypts. Chemical gradients applied to the basal-luminal axis compartmentalized the stem/progenitor cells and promoted appropriate lineage differentiation along the in vitro crypt axis so that the tissue possessed a crypt stem cell niche as well as a layer of differentiated cells covering the luminal surface. This new approach combining microengineered scaffolds, native chemical gradients, and biophysical cues to control primary epithelium ex vivo can serve as a highly functional and physiologically relevant in vitro tissue model.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Dulan B Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Peter J Attayek
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Mark I Reed
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Matthew DiSalvo
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Daniel L Nguyen
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Johanna S Dutton
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Michael S Lebhar
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Scott J Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Christopher E Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Scott T Magness
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Nancy L Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States.,Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27607, United States
| |
Collapse
|
55
|
Weng X, Neethirajan S. Ensuring food safety: Quality monitoring using microfluidics. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.04.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
56
|
|
57
|
A microengineered collagen scaffold for generating a polarized crypt-villus architecture of human small intestinal epithelium. Biomaterials 2017; 128:44-55. [PMID: 28288348 DOI: 10.1016/j.biomaterials.2017.03.005] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/04/2017] [Accepted: 03/04/2017] [Indexed: 02/06/2023]
Abstract
The human small intestinal epithelium possesses a distinct crypt-villus architecture and tissue polarity in which proliferative cells reside inside crypts while differentiated cells are localized to the villi. Indirect evidence has shown that the processes of differentiation and migration are driven in part by biochemical gradients of factors that specify the polarity of these cellular compartments; however, direct evidence for gradient-driven patterning of this in vivo architecture has been hampered by limitations of the in vitro systems available. Enteroid cultures are a powerful in vitro system; nevertheless, these spheroidal structures fail to replicate the architecture and lineage compartmentalization found in vivo, and are not easily subjected to gradients of growth factors. In the current work, we report the development of a micropatterned collagen scaffold with suitable extracellular matrix and stiffness to generate an in vitro self-renewing human small intestinal epithelium that replicates key features of the in vivo small intestine: a crypt-villus architecture with appropriate cell-lineage compartmentalization and an open and accessible luminal surface. Chemical gradients applied to the crypt-villus axis promoted the creation of a stem/progenitor-cell zone and supported cell migration along the crypt-villus axis. This new approach combining microengineered scaffolds, biophysical cues and chemical gradients to control the intestinal epithelium ex vivo can serve as a physiologically relevant mimic of the human small intestinal epithelium, and is broadly applicable to model other tissues that rely on gradients for physiological function.
Collapse
|
58
|
Fernandes JTS, Chutna O, Chu V, Conde JP, Outeiro TF. A Novel Microfluidic Cell Co-culture Platform for the Study of the Molecular Mechanisms of Parkinson's Disease and Other Synucleinopathies. Front Neurosci 2016; 10:511. [PMID: 27895548 PMCID: PMC5108800 DOI: 10.3389/fnins.2016.00511] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/25/2016] [Indexed: 01/09/2023] Open
Abstract
Although, the precise molecular mechanisms underlying Parkinson's disease (PD) are still elusive, it is now known that spreading of alpha-synuclein (aSyn) pathology and neuroinflammation are important players in disease progression. Here, we developed a novel microfluidic cell-culture platform for studying the communication between two different cell populations, a process of critical importance not only in PD but also in many biological processes. The integration of micro-valves in the device enabled us to control fluid routing, cellular microenvironments, and to simulate paracrine signaling. As proof of concept, two sets of experiments were designed to show how this platform can be used to investigate specific molecular mechanisms associated with PD. In one experiment, naïve H4 neuroglioma cells were co-cultured with cells expressing aSyn tagged with GFP (aSyn-GFP), to study the release and spreading of the protein. In our experimental set up, we induced the release of the contents of aSyn-GFP producing cells to the medium and monitored the protein's diffusion. In another experiment, H4 cells were co-cultured with N9 microglial cells to assess the interplay between two cell lines in response to environmental stimuli. Here, we observed an increase in the levels of reactive oxygen species in H4 cells cultured in the presence of activated N9 cells, confirming the cross talk between different cell populations. In summary, the platform developed in this study affords novel opportunities for the study of the molecular mechanisms involved in PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- João T S Fernandes
- Instituto de Engenharia de Sistemas E Computadores (INESC) - Microsistemas e Nanotecnologias and Institute of Nanoscience and Nanotechnology Lisbon, Portugal
| | - Oldriska Chutna
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa Lisbon, Portugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas E Computadores (INESC) - Microsistemas e Nanotecnologias and Institute of Nanoscience and Nanotechnology Lisbon, Portugal
| | - João P Conde
- Instituto de Engenharia de Sistemas E Computadores (INESC) - Microsistemas e Nanotecnologias and Institute of Nanoscience and NanotechnologyLisbon, Portugal; Departamento de Bioengenharia, Instituto Superior Técnico, Universidade de LisboaLisbon, Portugal
| | - Tiago F Outeiro
- Faculdade de Ciências Médicas, CEDOC - Chronic Diseases Research Center, Universidade Nova de LisboaLisbon, Portugal; Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center GöttingenGöttingen, Germany
| |
Collapse
|
59
|
Mortensen NP, Mercier KA, McRitchie S, Cavallo TB, Pathmasiri W, Stewart D, Sumner SJ. Microfluidics meets metabolomics to reveal the impact of Campylobacter jejuni infection on biochemical pathways. Biomed Microdevices 2016; 18:51. [PMID: 27231016 PMCID: PMC4939818 DOI: 10.1007/s10544-016-0076-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Microfluidic devices that are currently being used in pharmaceutical research also have a significant potential for utilization in investigating exposure to infectious agents. We have established a microfluidic device cultured with Caco-2 cells, and utilized metabolomics to investigate the biochemical responses to the bacterial pathogen Campylobacter jejuni. In the microfluidic devices, Caco-2 cells polarize at day 5, are uniform, have defined brush borders and tight junctions, and form a mucus layer. Metabolomics analysis of cell culture media collected from both Caco-2 cell culture systems demonstrated a more metabolic homogenous biochemical profile in the media collected from microfluidic devices, compared with media collected from transwells. GeneGo pathway mapping indicated that aminoacyl-tRNA biosynthesis was perturbed by fluid flow, suggesting that fluid dynamics and shear stress impacts the cells translational quality control. Both microfluidic device and transwell culturing systems were used to investigate the impact of Campylobacter jejuni infection on biochemical processes. Caco-2 cells cultured in either system were infected at day 5 with C. jejuni 81-176 for 48 h. Metabolomics analysis clearly differentiated C. jejuni 81-176 infected and non-infected medias collected from the microfluidic devices, and demonstrated that C. jejuni 81-176 infection in microfluidic devices impacts branched-chain amino acid metabolism, glycolysis, and gluconeogenesis. In contrast, no distinction was seen in the biochemical profiles of infected versus non-infected media collected from cells cultured in transwells. Microfluidic culturing conditions demonstrated a more metabolically homogenous cell population, and present the opportunity for studying host-pathogen interactions for extended periods of time.
Collapse
Affiliation(s)
- Ninell P Mortensen
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA.
| | - Kelly A Mercier
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Susan McRitchie
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Tammy B Cavallo
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Wimal Pathmasiri
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Delisha Stewart
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA
| | - Susan J Sumner
- Systems and Translational Sciences Discovery - Science - Technology, RTI International, 3040 Cornwallis Drive, Research Triangle Park, NC, 27709, USA.
- NIH Eastern Regional Comprehensive Metabolomics Resource Core, Systems and Translational Sciences, RTI International, 3040 East Cornwallis Road, Research Triangle Park, NC, 27709-2194, USA.
| |
Collapse
|
60
|
Jonczyk R, Kurth T, Lavrentieva A, Walter JG, Scheper T, Stahl F. Living Cell Microarrays: An Overview of Concepts. MICROARRAYS (BASEL, SWITZERLAND) 2016; 5:E11. [PMID: 27600077 PMCID: PMC5003487 DOI: 10.3390/microarrays5020011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023]
Abstract
Living cell microarrays are a highly efficient cellular screening system. Due to the low number of cells required per spot, cell microarrays enable the use of primary and stem cells and provide resolution close to the single-cell level. Apart from a variety of conventional static designs, microfluidic microarray systems have also been established. An alternative format is a microarray consisting of three-dimensional cell constructs ranging from cell spheroids to cells encapsulated in hydrogel. These systems provide an in vivo-like microenvironment and are preferably used for the investigation of cellular physiology, cytotoxicity, and drug screening. Thus, many different high-tech microarray platforms are currently available. Disadvantages of many systems include their high cost, the requirement of specialized equipment for their manufacture, and the poor comparability of results between different platforms. In this article, we provide an overview of static, microfluidic, and 3D cell microarrays. In addition, we describe a simple method for the printing of living cell microarrays on modified microscope glass slides using standard DNA microarray equipment available in most laboratories. Applications in research and diagnostics are discussed, e.g., the selective and sensitive detection of biomarkers. Finally, we highlight current limitations and the future prospects of living cell microarrays.
Collapse
Affiliation(s)
- Rebecca Jonczyk
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| | - Tracy Kurth
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| | - Johanna-Gabriela Walter
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| | - Thomas Scheper
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| | - Frank Stahl
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstr. 5, Hannover 30167, Germany.
| |
Collapse
|
61
|
Ramadan Q, Ting FCW. In vitro micro-physiological immune-competent model of the human skin. LAB ON A CHIP 2016; 16:1899-908. [PMID: 27098052 DOI: 10.1039/c6lc00229c] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Skin allergy, in particular, allergic contact dermatitis and irritant contact dermatitis, are common occupational and environmental health problems affecting the quality of life of a significant proportion of the world population. Since all new ingredients to be incorporated into a product are potential skin allergens, it is essential that these ingredients be first tested for their allergenic potential. However, despite the considerable effort using animal models to understand the underlying mechanism of skin sensitization, to date, the molecular and cellular responses due to skin contact with sensitizers are still not fully understood. To replace animal testing and to improve the prediction of skin sensitization, significant attention has been directed to the use of reconstructed organotypic in vitro models of human skin. Here we describe a miniaturized immune competent in vitro model of human skin based on 3D co-culture of immortalized human keratinocytes (HaCaT) as a model of the epidermis barrier and human leukemic monocyte lymphoma cell line (U937) as a model of human dendritic cells. The biological model was fitted in a microfluidic-based cell culture system that provides a dynamic cellular environment that mimics the in vivo environment of skin. The dynamic perfusion of culture media significantly improved the tight junction formation as evidenced by measuring higher values of TEER compared to static culture. This setting also maintained the high viability of cells over extended periods of time up to 17 days. The perfusion-based culture also allows growth of the cells at the air-liquid interface by exposing the apical side of the cells to air while providing the cell nutrients through a basolateral fluidic compartment. The microsystem has been evaluated to investigate the effect of the chemical and physical (UV irradiation) stimulation on the skin barrier (i.e. the TJ integrity). Three-tiered culture differential stimulation allowed the investigation of the role of the keratinocyte layer as a protection barrier to chemical/biological hazards.
Collapse
Affiliation(s)
- Qasem Ramadan
- Department of Bioelectronics, Institute of Microelectronics, Agency for Science, Technology and Research (A*STAR), Singapore.
| | | |
Collapse
|
62
|
Marx U, Andersson TB, Bahinski A, Beilmann M, Beken S, Cassee FR, Cirit M, Daneshian M, Fitzpatrick S, Frey O, Gaertner C, Giese C, Griffith L, Hartung T, Heringa MB, Hoeng J, de Jong WH, Kojima H, Kuehnl J, Luch A, Maschmeyer I, Sakharov D, Sips AJAM, Steger-Hartmann T, Tagle DA, Tonevitsky A, Tralau T, Tsyb S, van de Stolpe A, Vandebriel R, Vulto P, Wang J, Wiest J, Rodenburg M, Roth A. Biology-inspired microphysiological system approaches to solve the prediction dilemma of substance testing. ALTEX 2016; 33:272-321. [PMID: 27180100 PMCID: PMC5396467 DOI: 10.14573/altex.1603161] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023]
Abstract
The recent advent of microphysiological systems - microfluidic biomimetic devices that aspire to emulate the biology of human tissues, organs and circulation in vitro - is envisaged to enable a global paradigm shift in drug development. An extraordinary US governmental initiative and various dedicated research programs in Europe and Asia have led recently to the first cutting-edge achievements of human single-organ and multi-organ engineering based on microphysiological systems. The expectation is that test systems established on this basis would model various disease stages, and predict toxicity, immunogenicity, ADME profiles and treatment efficacy prior to clinical testing. Consequently, this technology could significantly affect the way drug substances are developed in the future. Furthermore, microphysiological system-based assays may revolutionize our current global programs of prioritization of hazard characterization for any new substances to be used, for example, in agriculture, food, ecosystems or cosmetics, thus, replacing laboratory animal models used currently. Thirty-six experts from academia, industry and regulatory bodies present here the results of an intensive workshop (held in June 2015, Berlin, Germany). They review the status quo of microphysiological systems available today against industry needs, and assess the broad variety of approaches with fit-for-purpose potential in the drug development cycle. Feasible technical solutions to reach the next levels of human biology in vitro are proposed. Furthermore, key organ-on-a-chip case studies, as well as various national and international programs are highlighted. Finally, a roadmap into the future is outlined, to allow for more predictive and regulatory-accepted substance testing on a global scale.
Collapse
|
63
|
Zheng F, Fu F, Cheng Y, Wang C, Zhao Y, Gu Z. Organ-on-a-Chip Systems: Microengineering to Biomimic Living Systems. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:2253-82. [PMID: 26901595 DOI: 10.1002/smll.201503208] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/09/2015] [Indexed: 05/20/2023]
Abstract
"Organ-on-a-chip" systems integrate microengineering, microfluidic technologies, and biomimetic principles to create key aspects of living organs faithfully, including critical microarchitecture, spatiotemporal cell-cell interactions, and extracellular microenvironments. This creative platform and its multiorgan integration recapitulating organ-level structures and functions can bring unprecedented benefits to a diversity of applications, such as developing human in vitro models for healthy or diseased organs, enabling the investigation of fundamental mechanisms in disease etiology and organogenesis, benefiting drug development in toxicity screening and target discovery, and potentially serving as replacements for animal testing. Recent advances in novel designs and examples for developing organ-on-a-chip platforms are reviewed. The potential for using this emerging technology in understanding human physiology including mechanical, chemical, and electrical signals with precise spatiotemporal controls are discussed. The current challenges and future directions that need to be pursued for these proof-of-concept studies are also be highlighted.
Collapse
Affiliation(s)
- Fuyin Zheng
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | - Fanfan Fu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | - Yao Cheng
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | - Chunyan Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, 210096, China
| |
Collapse
|
64
|
Blaauboer BJ, Boobis AR, Bradford B, Cockburn A, Constable A, Daneshian M, Edwards G, Garthoff JA, Jeffery B, Krul C, Schuermans J. Considering new methodologies in strategies for safety assessment of foods and food ingredients. Food Chem Toxicol 2016; 91:19-35. [PMID: 26939913 DOI: 10.1016/j.fct.2016.02.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/25/2016] [Indexed: 12/28/2022]
Abstract
Toxicology and safety assessment are changing and require new strategies for evaluating risk that are less depending on apical toxicity endpoints in animal models and relying more on knowledge of the mechanism of toxicity. This manuscript describes a number of developments that could contribute to this change and implement this in a stepwise roadmap that can be applied for the evaluation of food and food ingredients. The roadmap was evaluated in four case studies by using literature and existing data. This preliminary evaluation was shown to be useful. However, this experience should be extended by including examples where experimental work needs to be included. To further implement these new insights in toxicology and safety assessment for the area of food and food ingredients, the recommendation is that stakeholders take action in addressing gaps in our knowledge, e.g. with regard to the applicability of the roadmap for mixtures and food matrices. Further development of the threshold of toxicological concern is needed, as well as cooperation with other sectors where similar schemes are under development. Moreover, a more comprehensive evaluation of the roadmap, also including the identification of the need for in vitro experimental work is recommended.
Collapse
Affiliation(s)
- Bas J Blaauboer
- Utrecht University, Division of Toxicology, Institute for Risk Assessment Sciences, PO Box 80.177, 3508 TD, Utrecht, The Netherlands
| | - Alan R Boobis
- Imperial College London, Department of Medicine, Centre for Pharmacology & Therapeutics, London, W12 0NN, United Kingdom
| | - Bobbie Bradford
- Unilever, Safety & Environmental Assurance Centre, London, EC4Y 0DY, United Kingdom
| | - Andrew Cockburn
- University of Newcastle, Toxico-Logical Consulting Ltd, The Old Boiler House, Moor Place Park, Kettle Green Lane, Much Hadham, Hertfordshire, SG10 6AA, United Kingdom
| | - Anne Constable
- Nestlé Research Centre, Vers-Chez-les-Blanc, 1000, Lausanne 26, Switzerland
| | - Mardas Daneshian
- University of Konstanz, Center for Alternatives to Animal Testing-Europe CAAT-Europe, 78457, Konstanz, Germany
| | - Gareth Edwards
- Consultant, 63 Woodlands Road., Sonning Common, Reading, Berkshire, RG4 9TD, United Kingdom
| | | | - Brett Jeffery
- Mars, Global Chemical Food Safety Group, Slough, SL1 4JX, United Kingdom
| | - Cyrille Krul
- University of Applied Sciences, Research Centre Technology & Innovation, Dept. Innovative Testing in Life Sciences & Chemistry, PO Box 12011, 3501 AA, Utrecht, The Netherlands; TNO Healthy Living, PO box 360, 3700 AJ Zeist, The Netherlands
| | | |
Collapse
|
65
|
Ramadan Q, Jing L. Characterization of tight junction disruption and immune response modulation in a miniaturized Caco-2/U937 coculture-based in vitro model of the human intestinal barrier. Biomed Microdevices 2016; 18:11. [DOI: 10.1007/s10544-016-0035-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
66
|
Jie M, Li HF, Lin L, Zhang J, Lin JM. Integrated microfluidic system for cell co-culture and simulation of drug metabolism. RSC Adv 2016. [DOI: 10.1039/c6ra10407j] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We present a microfluidic integrator for cell cocultivation and simulation of pharmaceutical kinetic processes of oral drugs including intestinal absorption, liver metabolism, and anticancer activity.
Collapse
Affiliation(s)
- Mingsha Jie
- State Key Laboratory of Chemical Resource Engineering
- Beijing University of Chemical Technology
- Beijing 100029
- China
- Department of Chemistry
| | - Hai-Fang Li
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Luyao Lin
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Jie Zhang
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| | - Jin-Ming Lin
- Department of Chemistry
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
- Tsinghua University
- Beijing 100084
| |
Collapse
|
67
|
Li R, Lv X, Zhang X, Saeed O, Deng Y. Microfluidics for cell-cell interactions: A review. Front Chem Sci Eng 2015. [DOI: 10.1007/s11705-015-1550-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
68
|
Microfluidic Organ/Body-on-a-Chip Devices at the Convergence of Biology and Microengineering. SENSORS 2015; 15:31142-70. [PMID: 26690442 PMCID: PMC4721768 DOI: 10.3390/s151229848] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/16/2015] [Accepted: 12/04/2015] [Indexed: 12/24/2022]
Abstract
Recent advances in biomedical technologies are mostly related to the convergence of biology with microengineering. For instance, microfluidic devices are now commonly found in most research centers, clinics and hospitals, contributing to more accurate studies and therapies as powerful tools for drug delivery, monitoring of specific analytes, and medical diagnostics. Most remarkably, integration of cellularized constructs within microengineered platforms has enabled the recapitulation of the physiological and pathological conditions of complex tissues and organs. The so-called “organ-on-a-chip” technology, which represents a new avenue in the field of advanced in vitro models, with the potential to revolutionize current approaches to drug screening and toxicology studies. This review aims to highlight recent advances of microfluidic-based devices towards a body-on-a-chip concept, exploring their technology and broad applications in the biomedical field.
Collapse
|
69
|
Raasch M, Rennert K, Jahn T, Peters S, Henkel T, Huber O, Schulz I, Becker H, Lorkowski S, Funke H, Mosig A. Microfluidically supported biochip design for culture of endothelial cell layers with improved perfusion conditions. Biofabrication 2015; 7:015013. [DOI: 10.1088/1758-5090/7/1/015013] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
70
|
Ramadan Q, Gijs MAM. In vitro micro-physiological models for translational immunology. LAB ON A CHIP 2015; 15:614-36. [PMID: 25501670 DOI: 10.1039/c4lc01271b] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The immune system is a source of regulation of the human body and is key for its stable functioning. Animal models have been successfully used for many years to study human immunity and diseases and provided significant contributions to the development of powerful new therapies. However, such models inevitably display differences from the human metabolism and disease state and therefore may correlate poorly with the human conditions. This explains the interest for the use of in vitro models of human cells, which have better potential to assist in understanding the physiological events that characterize the immune response in humans. Microfluidic technologies offer great capabilities to create miniaturized in vivo-like physiological models that mimic tissue-tissue interactions and simulate the body metabolism in both the healthy and diseased states. The micro-scale features of these microfluidic systems allow positioning heterogeneous cellular cultures in close proximity to each other in a dynamic fluidic environment, thereby allowing efficient cell-cell interactions and effectively narrowing the gap between in vivo and in vitro conditions. Due to the relative simplicity of these systems, compared to animal models, it becomes possible to investigate cell signaling by monitoring the metabolites transported from one tissue to another in real time. This allows studying detailed physiological events and in consequence understanding the influence of metabolites on a specific tissue/organ function as well as on the healthy/diseased state modulation. Numerous in vitro models of human organs have been developed during the last few years, aiming to mimic as closely as possible the in vivo characteristics of such organs. This technology is still in its infancy, but is promised a bright future in industrial and medical applications. Here we review the recent literature, in which functional microphysiological models have been developed to mimic tissues and to explore multi-tissue interactions, focusing in particular on the study of immune reactions, inflammation and the development of diseases. Also, an outlook on the opportunities and issues for further translational development of functional in vitro models in immunology will be presented.
Collapse
Affiliation(s)
- Qasem Ramadan
- Bioelectronics Laboratory, Institute of Microelectronics, 11 Science Park II, Singapore 117685.
| | | |
Collapse
|
71
|
Boero C, Casulli MA, Olivo J, Foglia L, Orso E, Mazza M, Carrara S, De Micheli G. Design, development, and validation of an in-situ biosensor array for metabolite monitoring of cell cultures. Biosens Bioelectron 2014; 61:251-9. [DOI: 10.1016/j.bios.2014.05.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/07/2014] [Accepted: 05/13/2014] [Indexed: 11/16/2022]
|
72
|
Halldorsson S, Lucumi E, Gómez-Sjöberg R, Fleming RMT. Advantages and challenges of microfluidic cell culture in polydimethylsiloxane devices. Biosens Bioelectron 2014; 63:218-231. [PMID: 25105943 DOI: 10.1016/j.bios.2014.07.029] [Citation(s) in RCA: 572] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/03/2014] [Accepted: 07/12/2014] [Indexed: 02/06/2023]
Abstract
Culture of cells using various microfluidic devices is becoming more common within experimental cell biology. At the same time, a technological radiation of microfluidic cell culture device designs is currently in progress. Ultimately, the utility of microfluidic cell culture will be determined by its capacity to permit new insights into cellular function. Especially insights that would otherwise be difficult or impossible to obtain with macroscopic cell culture in traditional polystyrene dishes, flasks or well-plates. Many decades of heuristic optimization have gone into perfecting conventional cell culture devices and protocols. In comparison, even for the most commonly used microfluidic cell culture devices, such as those fabricated from polydimethylsiloxane (PDMS), collective understanding of the differences in cellular behavior between microfluidic and macroscopic culture is still developing. Moving in vitro culture from macroscopic culture to PDMS based devices can come with unforeseen challenges. Changes in device material, surface coating, cell number per unit surface area or per unit media volume may all affect the outcome of otherwise standard protocols. In this review, we outline some of the advantages and challenges that may accompany a transition from macroscopic to microfluidic cell culture. We focus on decisive factors that distinguish macroscopic from microfluidic cell culture to encourage a reconsideration of how macroscopic cell culture principles might apply to microfluidic cell culture.
Collapse
Affiliation(s)
- Skarphedinn Halldorsson
- Center for Systems Biology and Biomedical Center, University of Iceland, Sturlugata 8, Reykjavik, Iceland
| | - Edinson Lucumi
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Rafael Gómez-Sjöberg
- Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA, United States of America
| | - Ronan M T Fleming
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 avenue des Hauts-Fourneaux, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
73
|
Thuenauer R, Rodriguez-Boulan E, Römer W. Microfluidic approaches for epithelial cell layer culture and characterisation. Analyst 2014; 139:3206-18. [PMID: 24668405 PMCID: PMC4286366 DOI: 10.1039/c4an00056k] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In higher eukaryotes, epithelial cell layers line most body cavities and form selective barriers that regulate the exchange of solutes between compartments. In order to fulfil these functions, the cells assume a polarised architecture and maintain two distinct plasma membrane domains, the apical domain facing the lumen and the basolateral domain facing other cells and the extracellular matrix. Microfluidic biochips offer the unique opportunity to establish novel in vitro models of epithelia in which the in vivo microenvironment of epithelial cells is precisely reconstituted. In addition, analytical tools to monitor biologically relevant parameters can be directly integrated on-chip. In this review we summarise recently developed biochip designs for culturing epithelial cell layers. Since endothelial cell layers, which line blood vessels, have similar barrier functions and polar organisation as epithelial cell layers, we also discuss biochips for culturing endothelial cell layers. Furthermore, we review approaches to integrate tools to analyse and manipulate epithelia and endothelia in microfluidic biochips; including methods to perform electrical impedance spectroscopy; methods to detect substances undergoing trans-epithelial transport via fluorescence, spectrophotometry, and mass spectrometry; techniques to mechanically stimulate cells via stretching and fluid flow-induced shear stress; and methods to carry out high-resolution imaging of vesicular trafficking using light microscopy. Taken together, this versatile microfluidic toolbox enables novel experimental approaches to characterise epithelial monolayers.
Collapse
Affiliation(s)
- Roland Thuenauer
- Institute of Biology II, Albert-Ludwigs-University Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany.
| | | | | |
Collapse
|
74
|
Three dimensional human small intestine models for ADME-Tox studies. Drug Discov Today 2014; 19:1587-94. [PMID: 24853950 DOI: 10.1016/j.drudis.2014.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/04/2014] [Accepted: 05/07/2014] [Indexed: 10/25/2022]
Abstract
In vitro human small intestine models play a crucial part in preclinical drug development. Although conventional 2D systems possess many advantages, such as facile accessibility and high-throughput capability, they can also provide misleading results due to their relatively poor recapitulation of in vivo physiology. Significant progress has recently been made in developing 3D human small intestine models, suggesting that more-reliable preclinical results could be obtained by recreating the 3D intestinal microenvironment in vitro. Although there are still many challenges, 3D human small intestine models have the potential to facilitate drug screening and drug development.
Collapse
|
75
|
Wang Y, Ahmad AA, Sims CE, Magness ST, Allbritton NL. In vitro generation of colonic epithelium from primary cells guided by microstructures. LAB ON A CHIP 2014; 14:1622-31. [PMID: 24647645 PMCID: PMC4037563 DOI: 10.1039/c3lc51353j] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The proliferative compartment of the colonic epithelium in vivo is located in the basal crypt where colonic stem cells and transit-amplifying cells reside and fuel the rapid renewal of non-proliferative epithelial cells as they migrate toward the gut lumen. To mimic this tissue polarity, microstructures composed of polydimethylsiloxane (PDMS) microwells and Matrigel micropockets were used to guide a combined 2-dimensional (2D) and 3-dimensional (3D) hybrid culture of primary crypts isolated from the murine colon. The 2D and 3D culture of crypts on a planar PDMS surface was first investigated in terms of cell proliferation and stem cell activity. 3D culture of crypts with overlaid Matrigel generated enclosed, but highly proliferative spheroids (termed colonoids). 2D culture of crypts produced a spreading monolayer of cells, which were non-proliferative. A combined 2D/3D hybrid culture was generated in a PDMS microwell platform on which crypts were loaded by centrifugation into microwells (diameter = 150 μm, depth = 150 μm) followed by addition of Matrigel that formed micropockets locking the crypts within the microwells. Embedded crypts first underwent 3D expansion inside the wells. After the cells filled the microwells, they migrated onto the surrounding surface forming a 2D monolayer in the array regions without Matrigel. This unique 2D/3D hybrid culture generated a continuous, millimeter-scale colonic epithelial tissue in vitro, which resembled the polarized architecture (i.e. distinct proliferative and non-proliferative zones) and geometry of the colonic epithelium in vivo. This work initiates the construction of a "colon-on-a-chip" using primary cells/tissues with the ultimate goal of producing the physiologic structure and organ-level function of the colon.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | |
Collapse
|
76
|
Foulke-Abel J, In J, Kovbasnjuk O, Zachos NC, Ettayebi K, Blutt SE, Hyser JM, Zeng XL, Crawford SE, Broughman JR, Estes MK, Donowitz M. Human enteroids as an ex-vivo model of host-pathogen interactions in the gastrointestinal tract. Exp Biol Med (Maywood) 2014; 239:1124-34. [PMID: 24719375 DOI: 10.1177/1535370214529398] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Currently, 9 out of 10 experimental drugs fail in clinical studies. This has caused a 40% plunge in the number of drugs approved by the US Food and Drug Administration (FDA) since 2005. It has been suggested that the mechanistic differences between human diseases modeled in animals (mostly rodents) and the pathophysiology of human diseases might be one of the critical factors that contribute to drug failure in clinical trials. Rapid progress in the field of human stem cell technology has allowed the in-vitro recreation of human tissue that should complement and expand upon the limitations of cell and animal models currently used to study human diseases and drug toxicity. Recent success in the identification and isolation of human intestinal epithelial stem cells (Lgr5(+)) from the small intestine and colon has led to culture of functional intestinal epithelial units termed organoids or enteroids. Intestinal enteroids are comprised of all four types of normal epithelial cells and develop a crypt-villus differentiation axis. They demonstrate major intestinal physiologic functions, including Na(+) absorption and Cl(-) secretion. This review discusses the recent progress in establishing human enteroids as a model of infectious diarrheal diseases such as cholera, rotavirus, and enterohemorrhagic Escherichia coli, and use of the enteroids to determine ways to correct the diarrhea-induced ion transport abnormalities via drug therapy.
Collapse
Affiliation(s)
- Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Julie In
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Olga Kovbasnjuk
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Khalil Ettayebi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sarah E Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sue E Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R Broughman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
77
|
Giese C, Marx U. Human immunity in vitro - solving immunogenicity and more. Adv Drug Deliv Rev 2014; 69-70:103-22. [PMID: 24447895 DOI: 10.1016/j.addr.2013.12.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 12/24/2022]
Abstract
It has been widely recognised that the phylogenetic distance between laboratory animals and humans limits the former's predictive value for immunogenicity testing of biopharmaceuticals and nanostructure-based drug delivery and adjuvant systems. 2D in vitro assays have been established in conventional culture plates with little success so far. Here, we detail the status of various 3D approaches to emulate innate immunity in non-lymphoid organs and adaptive immune response in human professional lymphoid immune organs in vitro. We stress the tight relationship between the necessarily changing architecture of professional lymphoid organs at rest and when activated by pathogens, and match it with the immunity identified in vitro. Recommendations for further improvements of lymphoid tissue architecture relevant to the development of a sustainable adaptive immune response in vitro are summarized. In the end, we sketch a forecast of translational innovations in the field to model systemic innate and adaptive immunity in vitro.
Collapse
Affiliation(s)
| | - Uwe Marx
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| |
Collapse
|
78
|
Polini A, Prodanov L, Bhise NS, Manoharan V, Dokmeci MR, Khademhosseini A. Organs-on-a-chip: a new tool for drug discovery. Expert Opin Drug Discov 2014; 9:335-52. [PMID: 24620821 DOI: 10.1517/17460441.2014.886562] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The development of emerging in vitro tissue culture platforms can be useful for predicting human response to new compounds, which has been traditionally challenging in the field of drug discovery. Recently, several in vitro tissue-like microsystems, also known as 'organs-on-a-chip', have emerged to provide new tools for better evaluating the effects of various chemicals on human tissue. AREAS COVERED The aim of this article is to provide an overview of the organs-on-a-chip systems that have been recently developed. First, the authors introduce single-organ platforms, focusing on the most studied organs such as liver, heart, blood vessels and lung. Later, the authors briefly describe tumor-on-a-chip platforms and highlight their application for testing anti-cancer drugs. Finally, the article reports a few examples of other organs integrated in microfluidic chips along with preliminary multiple-organs-on-a-chip examples. The article also highlights key fabrication points as well as the main application areas of these devices. EXPERT OPINION This field is still at an early stage and major challenges need to be addressed prior to the embracement of these technologies by the pharmaceutical industry. To produce predictive drug screening platforms, several organs have to be integrated into a single microfluidic system representative of a humanoid. The routine production of metabolic biomarkers of the organ constructs, as well as their physical environment, have to be monitored prior to and during the delivery of compounds of interest to be able to translate the findings into useful discoveries.
Collapse
Affiliation(s)
- Alessandro Polini
- Brigham and Women's Hospital, Harvard Medical School, Division of Biomedical Engineering, Department of Medicine , Cambridge, MA 02139 , USA
| | | | | | | | | | | |
Collapse
|
79
|
Luni C, Serena E, Elvassore N. Human-on-chip for therapy development and fundamental science. Curr Opin Biotechnol 2014; 25:45-50. [DOI: 10.1016/j.copbio.2013.08.015] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 08/22/2013] [Indexed: 12/21/2022]
|
80
|
Huang C, Ramadan Q, Wacker JB, Tekin HC, Ruffert C, Vergères G, Silacci P, Gijs MAM. Microfluidic chip for monitoring Ca2+transport through a confluent layer of intestinal cells. RSC Adv 2014. [DOI: 10.1039/c4ra09370d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Microfluidic-based Caco-2 culture model provides a useful tool for investigating of metabolites transport and nutrikinetics studies.
Collapse
Affiliation(s)
- C. Huang
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| | - Q. Ramadan
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| | - J. B. Wacker
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| | - H. C. Tekin
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| | - C. Ruffert
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| | - G. Vergères
- Institute for Food Science
- Agroscope
- Federal Office of Agriculture
- Berne, Switzerland
| | - P. Silacci
- Institute for Food Science
- Agroscope
- Federal Office of Agriculture
- Berne, Switzerland
| | - M. A. M. Gijs
- Laboratory of Microsystems
- École Polytechnique Fédérale de Lausanne
- Switzerland
| |
Collapse
|
81
|
Wang Y, Ahmad AA, Shah PK, Sims CE, Magness ST, Allbritton NL. Capture and 3D culture of colonic crypts and colonoids in a microarray platform. LAB ON A CHIP 2013; 13:4625-34. [PMID: 24113577 PMCID: PMC3841105 DOI: 10.1039/c3lc50813g] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Crypts are the basic structural and functional units of colonic epithelium and can be isolated from the colon and cultured in vitro into multi-cell spheroids termed "colonoids". Both crypts and colonoids are ideal building blocks for construction of an in vitro tissue model of the colon. Here we proposed and tested a microengineered platform for capture and in vitro 3D culture of colonic crypts and colonoids. An integrated platform was fabricated from polydimethylsiloxane which contained two fluidic layers separated by an array of cylindrical microwells (150 μm diameter, 150 μm depth) with perforated bottoms (30 μm opening, 10 μm depth) termed "microstrainers". As fluid moved through the array, crypts or colonoids were retained in the microstrainers with a >90% array-filling efficiency. Matrigel as an extracellular matrix was then applied to the microstrainers to generate isolated Matrigel pockets encapsulating the crypts or colonoids. After supplying the essential growth factors, epidermal growth factor, Wnt-3A, R-spondin 2 and noggin, 63 ± 13% of the crypts and 77 ± 8% of the colonoids cultured in the microstrainers over a 48-72 h period formed viable 3D colonoids. Thus colonoid growth on the array was similar to that under standard culture conditions (78 ± 5%). Additionally the colonoids displayed the same morphology and similar numbers of stem and progenitor cells as those under standard culture conditions. Immunofluorescence staining confirmed that the differentiated cell-types of the colon, goblet cells, enteroendocrine cells and absorptive enterocytes, formed on the array. To demonstrating the utility of the array in tracking the colonoid fate, quantitative fluorescence analysis was performed on the arrayed colonoids exposed to reagents such as Wnt-3A and the γ-secretase inhibitor LY-411575. The successful formation of viable, multi-cell type colonic tissue on the microengineered platform represents a first step in the building of a "colon-on-a-chip" with the goal of producing the physiologic structure and organ-level function of the colon for controlled experiments.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | | | | | | | |
Collapse
|
82
|
Ghaye J, Kamat MA, Corbino-Giunta L, Silacci P, Vergères G, De Micheli G, Carrara S. Image thresholding techniques for localization of sub-resolution fluorescent biomarkers. Cytometry A 2013; 83:1001-16. [PMID: 24105983 DOI: 10.1002/cyto.a.22345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 04/29/2013] [Accepted: 07/16/2013] [Indexed: 11/09/2022]
Abstract
In this article, we explore adaptive global and local segmentation techniques for a lab-on-chip nutrition monitoring system (NutriChip). The experimental setup consists of Caco-2 intestinal cells that can be artificially stimulated to trigger an immune response. The eventual response is optically monitored using immunofluoresence techniques targeting toll-like receptor 2 (TLR2). Two problems of interest need to be addressed by means of image processing. First, a new cell sample must be properly classified as stimulated or not. Second, the location of the stained TLR2 must be recovered in case the sample has been stimulated. The algorithmic approach to solving these problems is based on the ability of a segmentation technique to properly segment fluorescent spots. The sample classification is based on the amount and intensity of the segmented pixels, while the various segmenting blobs provide an approximate localization of TLR2. A novel local thresholding algorithm and three well-known spot segmentation techniques are compared in this study. Quantitative assessment of these techniques based on real and synthesized data demonstrates the improved segmentation capabilities of the proposed algorithm.
Collapse
Affiliation(s)
- Julien Ghaye
- Laboratory of Integrated Systems (LSI), Swiss Federal Institute of Technology, EPFL, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
83
|
Hitzbleck M, Delamarche E. Reagents in microfluidics: an 'in' and 'out' challenge. Chem Soc Rev 2013; 42:8494-516. [PMID: 23925517 DOI: 10.1039/c3cs60118h] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Microfluidic devices are excellent at downscaling chemical and biochemical reactions and thereby can make reactions faster, better and more efficient. It is therefore understandable that we are seeing these devices being developed and used for many applications and research areas. However, microfluidic devices are more complex than test tubes or microtitre plates and the integration of reagents into them is a real challenge. This review looks at state-of-the-art methods and strategies for integrating various classes of reagents inside microfluidics and similarly surveys how reagents can be released inside microfluidics. The number of methods used for integrating and releasing reagents is surprisingly large and involves reagents in dry and liquid forms, directly-integrated reagents or reagents linked to carriers, as well as active, passive and hybrid release methods. We also made a brief excursion into the field of drug release and delivery. With this review, we hope to provide a large number of examples of integrating and releasing reagents that can be used by developers and users of microfluidics for their specific needs.
Collapse
|
84
|
Selimović S, Dokmeci MR, Khademhosseini A. Organs-on-a-chip for drug discovery. Curr Opin Pharmacol 2013; 13:829-33. [PMID: 23850526 DOI: 10.1016/j.coph.2013.06.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/14/2013] [Accepted: 06/17/2013] [Indexed: 11/17/2022]
Abstract
The current drug discovery process is arduous and costly, and a majority of the drug candidates entering clinical trials fail to make it to the marketplace. The standard static well culture approaches, although useful, do not fully capture the intricate in vivo environment. By merging the advances in microfluidics with microfabrication technologies, novel platforms are being introduced that lead to the creation of organ functions on a single chip. Within these platforms, microengineering enables precise control over the cellular microenvironment, whereas microfluidics provides an ability to perfuse the constructs on a chip and to connect individual sections with each other. This approach results in microsystems that may better represent the in vivo environment. These organ-on-a-chip platforms can be utilized for developing disease models as well as for conducting drug testing studies. In this article, we highlight several key developments in these microscale platforms for drug discovery applications.
Collapse
Affiliation(s)
- Seila Selimović
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | |
Collapse
|