101
|
Kim J, Lee H, Selimović Š, Gauvin R, Bae H. Organ-On-A-Chip: Development and Clinical Prospects Toward Toxicity Assessment with an Emphasis on Bone Marrow. Drug Saf 2015; 38:409-18. [DOI: 10.1007/s40264-015-0284-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
102
|
Abstract
The new cover of Experimental Biology and Medicine features the hermeneutic circle of biology, a concept we have adapted from the hermeneutic principle that one understands the whole only in terms of each part and the parts only in terms of the whole. Our hermeneutic circle summarizes the course of experimental biology through 2500 years of the achievements of reductionist research (understanding the parts), which culminates in our ability to rapidly sequence the genome. Rather than returning along the same path in a constructionist approach that simply builds upon this knowledge, but in reverse, an alternative is to close the circle with synthetic constructions that seek to integrate the full complexity of biological and physiological systems (understanding the whole), of which organs-on-chips are one example. This closing of the circle cannot be a comprehensively accurate representation of biology, but it can be a synthetic one that effectively defines particular biological subsystems. The illustration of the hermeneutic circle of biology is also intended to suggest both the multiple cycles that may be required to reach such a synthesis and the expansion of the circle in an outward spiral as knowledge increases. Our commentary explains the symbolism of the new cover in a philosophical and scientific discussion.
Collapse
Affiliation(s)
- John P Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics & Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| | - Andrew P Porter
- Center for Theology and the Natural Sciences, Graduate Theological Union, Berkeley, CA 94709-1212, USA
| |
Collapse
|
103
|
Zhang YS, Ribas J, Nadhman A, Aleman J, Selimović Š, Lesher-Perez SC, Wang T, Manoharan V, Shin SR, Damilano A, Annabi N, Dokmeci MR, Takayama S, Khademhosseini A. A cost-effective fluorescence mini-microscope for biomedical applications. LAB ON A CHIP 2015; 15:3661-3669. [PMID: 26282117 PMCID: PMC4550514 DOI: 10.1039/c5lc00666j] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
We have designed and fabricated a miniature microscope from off-the-shelf components and a webcam, with built-in fluorescence capability for biomedical applications. The mini-microscope was able to detect both biochemical parameters, such as cell/tissue viability (e.g. live/dead assay), and biophysical properties of the microenvironment such as oxygen levels in microfabricated tissues based on an oxygen-sensitive fluorescent dye. This mini-microscope has adjustable magnifications from 8-60×, achieves a resolution as high as <2 μm, and possesses a long working distance of 4.5 mm (at a magnification of 8×). The mini-microscope was able to chronologically monitor cell migration and analyze beating of microfluidic liver and cardiac bioreactors in real time, respectively. The mini-microscope system is cheap, and its modularity allows convenient integration with a wide variety of pre-existing platforms including, but not limited to, cell culture plates, microfluidic devices, and organs-on-a-chip systems. Therefore, we envision its widespread application in cell biology, tissue engineering, biosensing, microfluidics, and organs-on-chips, which can potentially replace conventional bench-top microscopy where long-term in situ and large-scale imaging/analysis is required.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Goers L, Freemont P, Polizzi KM. Co-culture systems and technologies: taking synthetic biology to the next level. J R Soc Interface 2014; 11:rsif.2014.0065. [PMID: 24829281 DOI: 10.1098/rsif.2014.0065] [Citation(s) in RCA: 374] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Co-culture techniques find myriad applications in biology for studying natural or synthetic interactions between cell populations. Such techniques are of great importance in synthetic biology, as multi-species cell consortia and other natural or synthetic ecology systems are widely seen to hold enormous potential for foundational research as well as novel industrial, medical and environmental applications with many proof-of-principle studies in recent years. What is needed for co-cultures to fulfil their potential? Cell-cell interactions in co-cultures are strongly influenced by the extracellular environment, which is determined by the experimental set-up, which therefore needs to be given careful consideration. An overview of existing experimental and theoretical co-culture set-ups in synthetic biology and adjacent fields is given here, and challenges and opportunities involved in such experiments are discussed. Greater focus on foundational technology developments for co-cultures is needed for many synthetic biology systems to realize their potential in both applications and answering biological questions.
Collapse
Affiliation(s)
- Lisa Goers
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK Centre for Synthetic Biology and Innovation, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Paul Freemont
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK Centre for Synthetic Biology and Innovation, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Karen M Polizzi
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK Centre for Synthetic Biology and Innovation, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|
105
|
Moraes C. Micro, soft, windows: integrating super-resolution viewing capabilities into soft lithographic devices. Integr Biol (Camb) 2014; 7:10-3. [PMID: 25514253 DOI: 10.1039/c4ib90046d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Microengineered cell culture environments afford experimentalists with the critical ability to study cells in precisely-defined, yet physiologically-realistic environments. A significant, but often overlooked, feature of these technologies is the unique ability to optically probe cellular and sub-cellular processes during culture in these complex environments, thereby obtaining information that would not be possible via conventional techniques. Motivated by the recent presentation of the Nobel prizes for super-resolution imaging and more recent technological breakthroughs in lattice-based light sheet microscopy, in this research highlight we survey recent innovations in the design of microfluidic cell culture platforms, that will ultimately allow experimentalists to probe biological activity with high-spatial and temporal-resolution. These advances will provide new technology-driven windows into biological processes and mechanisms.
Collapse
Affiliation(s)
- Christopher Moraes
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada.
| |
Collapse
|
106
|
Buchmann A, Alber M, Zartman JJ. Sizing it up: The mechanical feedback hypothesis of organ growth regulation. Semin Cell Dev Biol 2014; 35:73-81. [DOI: 10.1016/j.semcdb.2014.06.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/26/2014] [Indexed: 11/28/2022]
|
107
|
Penco S, Venco E, Lio A. Lost in translation: the need for better tools. Altern Lab Anim 2014; 42:P41-5. [PMID: 25290949 DOI: 10.1177/026119291404200411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although for most pharmaceutical compounds the final aim is improving human health, almost all the methods used to identify and pursue therapeutic targets and to obtain new potential drugs have traditionally focused on animal models
Collapse
|
108
|
Zhu X, Bouffanais R, Yue DKP. Persistent cellular motion control and trapping using mechanotactic signaling. PLoS One 2014; 9:e105406. [PMID: 25207940 PMCID: PMC4160188 DOI: 10.1371/journal.pone.0105406] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/19/2014] [Indexed: 11/19/2022] Open
Abstract
Chemotactic signaling and the associated directed cell migration have been extensively studied owing to their importance in emergent processes of cellular aggregation. In contrast, mechanotactic signaling has been relatively overlooked despite its potential for unique ways to artificially signal cells with the aim to effectively gain control over their motile behavior. The possibility of mimicking cellular mechanotactic signals offers a fascinating novel strategy to achieve targeted cell delivery for in vitro tissue growth if proven to be effective with mammalian cells. Using (i) optimal level of extracellular calcium ([Ca2+ ]ext mM) we found, (ii) controllable fluid shear stress of low magnitude (), and (iii) the ability to swiftly reverse flow direction (within one second), we are able to successfully signal Dictyostelium discoideum amoebae and trigger migratory responses with heretofore unreported control and precision. Specifically, we are able to systematically determine the mechanical input signal required to achieve any predetermined sequences of steps including straightforward motion, reversal and trapping. The mechanotactic cellular trapping is achieved for the first time and is associated with a stalling frequency of Hz for a reversing direction mechanostimulus, above which the cells are effectively trapped while maintaining a high level of directional sensing. The value of this frequency is very close to the stalling frequency recently reported for chemotactic cell trapping [Meier B, et al. (2011) Proc Natl Acad Sci USA 108:11417–11422], suggesting that the limiting factor may be the slowness of the internal chemically-based motility apparatus.
Collapse
Affiliation(s)
- Xiaoying Zhu
- Singapore University of Technology and Design, Singapore, Singapore
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Roland Bouffanais
- Singapore University of Technology and Design, Singapore, Singapore
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| | - Dick K. P. Yue
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| |
Collapse
|
109
|
Wikswo JP. The relevance and potential roles of microphysiological systems in biology and medicine. Exp Biol Med (Maywood) 2014; 239:1061-72. [PMID: 25187571 PMCID: PMC4330974 DOI: 10.1177/1535370214542068] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Microphysiological systems (MPS), consisting of interacting organs-on-chips or tissue-engineered, 3D organ constructs that use human cells, present an opportunity to bring new tools to biology, medicine, pharmacology, physiology, and toxicology. This issue of Experimental Biology and Medicine describes the ongoing development of MPS that can serve as in-vitro models for bone and cartilage, brain, gastrointestinal tract, lung, liver, microvasculature, reproductive tract, skeletal muscle, and skin. Related topics addressed here are the interconnection of organs-on-chips to support physiologically based pharmacokinetics and drug discovery and screening, and the microscale technologies that regulate stem cell differentiation. The initial motivation for creating MPS was to increase the speed, efficiency, and safety of pharmaceutical development and testing, paying particular regard to the fact that neither monolayer monocultures of immortal or primary cell lines nor animal studies can adequately recapitulate the dynamics of drug-organ, drug-drug, and drug-organ-organ interactions in humans. Other applications include studies of the effect of environmental toxins on humans, identification, characterization, and neutralization of chemical and biological weapons, controlled studies of the microbiome and infectious disease that cannot be conducted in humans, controlled differentiation of induced pluripotent stem cells into specific adult cellular phenotypes, and studies of the dynamics of metabolism and signaling within and between human organs. The technical challenges are being addressed by many investigators, and in the process, it seems highly likely that significant progress will be made toward providing more physiologically realistic alternatives to monolayer monocultures or whole animal studies. The effectiveness of this effort will be determined in part by how easy the constructs are to use, how well they function, how accurately they recapitulate and report human pharmacology and toxicology, whether they can be generated in large numbers to enable parallel studies, and if their use can be standardized consistent with the practices of regulatory science.
Collapse
Affiliation(s)
- John P Wikswo
- Departments of Biomedical Engineering, Molecular Physiology and Biophysics, and Physics and Astronomy, Vanderbilt University, The Vanderbilt Institute for Integrative Biosystems Research and Education, VU Station B 351807, Nashville, TN 37235-1807, USA
| |
Collapse
|
110
|
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: 3.2] [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
|
111
|
Sung JH, Srinivasan B, Esch MB, McLamb WT, Bernabini C, Shuler ML, Hickman JJ. Using physiologically-based pharmacokinetic-guided "body-on-a-chip" systems to predict mammalian response to drug and chemical exposure. Exp Biol Med (Maywood) 2014; 239:1225-39. [PMID: 24951471 DOI: 10.1177/1535370214529397] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The continued development of in vitro systems that accurately emulate human response to drugs or chemical agents will impact drug development, our understanding of chemical toxicity, and enhance our ability to respond to threats from chemical or biological agents. A promising technology is to build microscale replicas of humans that capture essential elements of physiology, pharmacology, and/or toxicology (microphysiological systems). Here, we review progress on systems for microscale models of mammalian systems that include two or more integrated cellular components. These systems are described as a "body-on-a-chip", and utilize the concept of physiologically-based pharmacokinetic (PBPK) modeling in the design. These microscale systems can also be used as model systems to predict whole-body responses to drugs as well as study the mechanism of action of drugs using PBPK analysis. In this review, we provide examples of various approaches to construct such systems with a focus on their physiological usefulness and various approaches to measure responses (e.g. chemical, electrical, or mechanical force and cellular viability and morphology). While the goal is to predict human response, other mammalian cell types can be utilized with the same principle to predict animal response. These systems will be evaluated on their potential to be physiologically accurate, to provide effective and efficient platform for analytics with accessibility to a wide range of users, for ease of incorporation of analytics, functional for weeks to months, and the ability to replicate previously observed human responses.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Chemical Engineering, Hongik University, Seoul 121-791, Republic of Korea
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Mandy Brigitte Esch
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - William T McLamb
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Catia Bernabini
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32826, USA Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32816, USA
| |
Collapse
|
112
|
Dixon AR, Moraes C, Csete ME, Thouless MD, Philbert MA, Takayama S. One-dimensional patterning of cells in silicone wells via compression-induced fracture. J Biomed Mater Res A 2014; 102:1361-9. [PMID: 23733484 PMCID: PMC3912204 DOI: 10.1002/jbm.a.34814] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/04/2013] [Accepted: 05/21/2013] [Indexed: 12/13/2022]
Abstract
We have adapted our existing compression-induced fracture technology to cell culture studies by generating linear patterns on a complex cell culture well structure rather than on simple solid constructs. We present a simple method to create one-dimensional (1D), submicron, and linear patterns of extracellular matrix on a multilayer silicone material. We identified critical design parameters necessary to optimize compression-induced fracture patterning on the wells, and applied stresses using compression Hoffman clamps. Finite-element analyses show that the incorporation of the well improves stress homogeneity (stress variation = 25%), and, thus, crack uniformity over the patterned region. Notably, a shallow well with a thick base (vs. deeper wells with thinner bases) reduces out-of-plane deflections by greater than a sixth in the cell culture region, improving clarity for optical imaging. The comparison of cellular and nuclear shape indices of a neuroblast line cultured on patterned 1D lines and unpatterned 2D surfaces reveals significant differences in cellular morphology, which could impact many cellular functions. Because 1D cell cultures recapitulate many important phenotypical traits of 3D cell cultures, our culture system offers a simple means to further study the relationship between 1D and 3D cell culture environments, without demanding expensive engineering techniques and expertise.
Collapse
Affiliation(s)
- Angela R. Dixon
- Toxicology Program, School of Public Health, University of Michigan, Ann Arbor, Michigan
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor Michigan
| | - Christopher Moraes
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor Michigan
| | - Marie E. Csete
- Departments of Anesthesiology and Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan (Current affiliation: AABB Center for Cellular Therapies, Bethesda, Maryland)
| | - M. D. Thouless
- Departments of Mechanical Engineering and Materials Science & Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan
| | - Martin A. Philbert
- Toxicology Program, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Shuichi Takayama
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor Michigan
- Macromolecular Science and Engineering Program, College of Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
113
|
Kim D, Wu X, Young AT, Haynes CL. Microfluidics-based in vivo mimetic systems for the study of cellular biology. Acc Chem Res 2014; 47:1165-73. [PMID: 24555566 PMCID: PMC3993883 DOI: 10.1021/ar4002608] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
The human body is a complex network of molecules,
organelles, cells,
tissues, and organs: an uncountable number of interactions and transformations
interconnect all the system’s components. In addition to these
biochemical components, biophysical components, such as pressure,
flow, and morphology, and the location of all of these interactions
play an important role in the human body. Technical difficulties have
frequently limited researchers from observing cellular biology as
it occurs within the human body, but some state-of-the-art analytical
techniques have revealed distinct cellular behaviors that occur only
in the context of the interactions. These types of findings have inspired
bioanalytical chemists to provide new tools to better understand these
cellular behaviors and interactions. What blocks us from understanding
critical biological interactions
in the human body? Conventional approaches are often too naïve
to provide realistic data and in vivo whole animal studies give complex
results that may or may not be relevant for humans. Microfluidics
offers an opportunity to bridge these two extremes: while these studies
will not model the complexity of the in vivo human system, they can
control the complexity so researchers can examine critical factors
of interest carefully and quantitatively. In addition, the use of
human cells, such as cells isolated from donated blood, captures human-relevant
data and limits the use of animals in research. In addition, researchers
can adapt these systems easily and cost-effectively to a variety of
high-end signal transduction mechanisms, facilitating high-throughput
studies that are also spatially, temporally, or chemically resolved.
These strengths should allow microfluidic platforms to reveal critical
parameters in the human body and provide insights that will help with
the translation of pharmacological advances to clinical trials. In this Account, we describe selected microfluidic innovations
within the last 5 years that focus on modeling both biophysical and
biochemical interactions in cellular communication, such as flow and
cell–cell networks. We also describe more advanced systems
that mimic higher level biological networks, such as organ on-a-chip
and animal on-a-chip models. Since the first papers in the early 1990s,
interest in the bioanalytical use of microfluidics has grown significantly.
Advances in micro-/nanofabrication technology have allowed researchers
to produce miniaturized, biocompatible assay platforms suitable for
microfluidic studies in biochemistry and chemical biology. Well-designed
microfluidic platforms can achieve quick, in vitro analyses on pico-
and femtoliter volume samples that are temporally, spatially, and
chemically resolved. In addition, controlled cell culture techniques
using a microfluidic platform have produced biomimetic systems that
allow researchers to replicate and monitor physiological interactions.
Pioneering work has successfully created cell–fluid, cell–cell,
cell–tissue, tissue–tissue, even organ-like level interfaces.
Researchers have monitored cellular behaviors in these biomimetic
microfluidic environments, producing validated model systems to understand
human pathophysiology and to support the development of new therapeutics.
Collapse
Affiliation(s)
- Donghyuk Kim
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street Southeast, Minneapolis, Minnesota 55455, United States
| | - Xiaojie Wu
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street Southeast, Minneapolis, Minnesota 55455, United States
| | - Ashlyn T. Young
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street Southeast, Minneapolis, Minnesota 55455, United States
| | - Christy L. Haynes
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street Southeast, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
114
|
Young EWK. Cells, tissues, and organs on chips: challenges and opportunities for the cancer tumor microenvironment. Integr Biol (Camb) 2014; 5:1096-109. [PMID: 23799587 DOI: 10.1039/c3ib40076j] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transition to increasingly sophisticated microfluidic systems has led to the emergence of "organ-on-chip" technology that can faithfully recapitulate organ-level function. Given the rapid progress at the interface between microfluidics and cell biology, there is need to provide a focused evaluation of the state-of-the-art in microfluidic systems for cancer research to advance development, accelerate discovery of novel insights, and facilitate cooperation between engineers, biologists and oncologists in the clinic. Here, we provide a focused review of microfluidics technology from cells- and tissues- to organs-on-chips with application toward studying the tumor microenvironment. Key aspects of the tumor microenvironment including angiogenesis, hypoxia, biochemical gradients, tumor-stromal interactions, and the extracellular matrix are summarized for both solid tumors and non-solid hematologic malignancies. An overview of microfluidic systems designed specifically to answer questions related to different aspects of the tumor microenvironment is provided, followed by an examination of how these systems offer new opportunities to study outstanding challenges related to the major cancer hallmarks. Challenges also remain for microfluidics engineers, but it is hoped that cooperation between engineers and biologists at the intersection of their respective fields will lead to significant impact on the utility of organs-on-chips in cancer research.
Collapse
Affiliation(s)
- Edmond W K Young
- Department of Mechanical & Industrial Engineering, University of Toronto, 5 King's College Road, MC314B, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
115
|
Moraes C, Labuz JM, Leung BM, Inoue M, Chun TH, Takayama S. On being the right size: scaling effects in designing a human-on-a-chip. Integr Biol (Camb) 2014; 5:1149-61. [PMID: 23925524 DOI: 10.1039/c3ib40040a] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Developing a human-on-a-chip by connecting multiple model organ systems would provide an intermediate screen for therapeutic efficacy and toxic side effects of drugs prior to conducting expensive clinical trials. However, correctly designing individual organs and scaling them relative to each other to make a functional microscale human analog is challenging, and a generalized approach has yet to be identified. In this work, we demonstrate the importance of rational design of both the individual organ and its relationship with other organs, using a simple two-compartment system simulating insulin-dependent glucose uptake in adipose tissues. We demonstrate that inter-organ scaling laws depend on both the number of cells and the spatial arrangement of those cells within the microfabricated construct. We then propose a simple and novel inter-organ 'metabolically supported functional scaling' approach predicated on maintaining in vivo cellular basal metabolic rates by limiting resources available to cells on the chip. This approach leverages findings from allometric scaling models in mammals that limited resources in vivo prompt cells to behave differently than in resource-rich in vitro cultures. Although applying scaling laws directly to tissues can result in systems that would be quite challenging to implement, engineering workarounds may be used to circumvent these scaling issues. Specific workarounds discussed include the limited oxygen carrying capacity of cell culture media when used as a blood substitute and the ability to engineer non-physiological structures to augment organ function, to create the transport-accessible, yet resource-limited environment necessary for cells to mimic in vivo functionality. Furthermore, designing the structure of individual tissues in each organ compartment may be a useful strategy to bypass scaling concerns at the inter-organ level.
Collapse
Affiliation(s)
- Christopher Moraes
- Department of Biomedical Engineering, College of Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
116
|
Yum K, Hong SG, Healy KE, Lee LP. Physiologically relevant organs on chips. Biotechnol J 2013; 9:16-27. [PMID: 24357624 DOI: 10.1002/biot.201300187] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 09/16/2013] [Accepted: 10/28/2013] [Indexed: 12/23/2022]
Abstract
Recent advances in integrating microengineering and tissue engineering have generated promising microengineered physiological models for experimental medicine and pharmaceutical research. Here we review the recent development of microengineered physiological systems, or also known as "ogans-on-chips", that reconstitute the physiologically critical features of specific human tissues and organs and their interactions. This technology uses microengineering approaches to construct organ-specific microenvironments, reconstituting tissue structures, tissue-tissue interactions and interfaces, and dynamic mechanical and biochemical stimuli found in specific organs, to direct cells to assemble into functional tissues. We first discuss microengineering approaches to reproduce the key elements of physiologically important, dynamic mechanical microenvironments, biochemical microenvironments, and microarchitectures of specific tissues and organs in microfluidic cell culture systems. This is followed by examples of microengineered individual organ models that incorporate the key elements of physiological microenvironments into single microfluidic cell culture systems to reproduce organ-level functions. Finally, microengineered multiple organ systems that simulate multiple organ interactions to better represent human physiology, including human responses to drugs, is covered in this review. This emerging organs-on-chips technology has the potential to become an alternative to 2D and 3D cell culture and animal models for experimental medicine, human disease modeling, drug development, and toxicology.
Collapse
Affiliation(s)
- Kyungsuk Yum
- Department of Bioengineering, University of California, Berkeley, CA, USA; Department of Materials Science and Engineering, University of Texas, Arlington, TX, USA
| | | | | | | |
Collapse
|
117
|
Young EWK. Advances in Microfluidic Cell Culture Systems for Studying Angiogenesis. ACTA ACUST UNITED AC 2013; 18:427-36. [DOI: 10.1177/2211068213495206] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
118
|
Design, fabrication and characterization of drug delivery systems based on lab-on-a-chip technology. Adv Drug Deliv Rev 2013; 65:1403-19. [PMID: 23726943 DOI: 10.1016/j.addr.2013.05.008] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 05/16/2013] [Accepted: 05/22/2013] [Indexed: 11/23/2022]
Abstract
Lab-on-a-chip technology is an emerging field evolving from the recent advances of micro- and nanotechnologies. The technology allows the integration of various components into a single microdevice. Microfluidics, the science and engineering of fluid flow in microscale, is the enabling underlying concept for lab-on-a-chip technology. The present paper reviews the design, fabrication and characterization of drug delivery systems based on this amazing technology. The systems are categorized and discussed according to the scales at which the drug is administered. Starting with the fundamentals on scaling laws of mass transfer and basic fabrication techniques, the paper reviews and discusses drug delivery devices for cellular, tissue and organism levels. At the cellular level, a concentration gradient generator integrated with a cell culture platform is the main drug delivery scheme of interest. At the tissue level, the synthesis of smart particles as drug carriers using lab-on-a-chip technology is the main focus of recent developments. At the organism level, microneedles and implantable devices with fluid-handling components are the main drug delivery systems. For drug delivery to a small organism that can fit into a microchip, devices similar to those of cellular level can be used.
Collapse
|
119
|
Tsui JH, Lee W, Pun SH, Kim J, Kim DH. Microfluidics-assisted in vitro drug screening and carrier production. Adv Drug Deliv Rev 2013; 65:1575-88. [PMID: 23856409 DOI: 10.1016/j.addr.2013.07.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/06/2013] [Accepted: 07/05/2013] [Indexed: 12/11/2022]
Abstract
Microfluidic platforms provide several unique advantages for drug development. In the production of drug carriers, physical properties such as size and shape, and chemical properties such as drug composition and pharmacokinetic parameters, can be modified simply and effectively by tuning the flow rate and geometries. Large numbers of carriers can then be fabricated with minimal effort and with little to no batch-to-batch variation. Additionally, cell or tissue culture models in microfluidic systems can be used as in vitro drug screening tools. Compared to in vivo animal models, microfluidic drug screening platforms allow for high-throughput and reproducible screening at a significantly lower cost, and when combined with current advances in tissue engineering, are also capable of mimicking native tissues. In this review, various microfluidic platforms for drug and gene carrier fabrication are reviewed to provide guidelines for designing appropriate carriers. In vitro microfluidic drug screening platforms designed for high-throughput analysis and replication of in vivo conditions are also reviewed to highlight future directions for drug research and development.
Collapse
Affiliation(s)
- Jonathan H Tsui
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
120
|
Li X, Mearns SM, Martins-Green M, Liu Y. Procedure for the development of multi-depth circular cross-sectional endothelialized microchannels-on-a-chip. J Vis Exp 2013:e50771. [PMID: 24193102 DOI: 10.3791/50771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Efforts have been focused on developing in vitro assays for the study of microvessels because in vivo animal studies are more time-consuming, expensive, and observation and quantification are very challenging. However, conventional in vitro microvessel assays have limitations when representing in vivo microvessels with respect to three-dimensional (3D) geometry and providing continuous fluid flow. Using a combination of photolithographic reflowable photoresist technique, soft lithography, and microfluidics, we have developed a multi-depth circular cross-sectional endothelialized microchannels-on-a-chip, which mimics the 3D geometry of in vivo microvessels and runs under controlled continuous perfusion flow. A positive reflowable photoresist was used to fabricate a master mold with a semicircular cross-sectional microchannel network. By the alignment and bonding of the two polydimethylsiloxane (PDMS) microchannels replicated from the master mold, a cylindrical microchannel network was created. The diameters of the microchannels can be well controlled. In addition, primary human umbilical vein endothelial cells (HUVECs) seeded inside the chip showed that the cells lined the inner surface of the microchannels under controlled perfusion lasting for a time period between 4 days to 2 weeks.
Collapse
Affiliation(s)
- Xiang Li
- Lane Department of Computer Science and Electrical Engineering, West Virginia University
| | | | | | | |
Collapse
|
121
|
Abstract
'Organs-on-chips' are microengineered biomimetic systems containing microfluidic channels lined by living human cells, which replicate key functional units of living organs to reconstitute integrated human organ-level pathophysiology in vitro. These microdevices can be used to test efficacy and toxicity of drugs and chemicals, and to create in vitro models of human disease. Thus, they potentially represent low-cost alternatives to conventional animal models for pharmaceutical, chemical and environmental applications. Here we describe a protocol for the fabrication, microengineering and operation of these microfluidic organ-on-chip systems. First, microengineering is used to fabricate a multilayered microfluidic device that contains two parallel elastomeric microchannels separated by a thin porous flexible membrane, along with two full-height, hollow vacuum chambers on either side; this requires ∼3.5 d to complete. To create a 'breathing' lung-on-a-chip that mimics the mechanically active alveolar-capillary interface of the living human lung, human alveolar epithelial cells and microvascular endothelial cells are cultured in the microdevice with physiological flow and cyclic suction applied to the side chambers to reproduce rhythmic breathing movements. We describe how this protocol can be easily adapted to develop other human organ chips, such as a gut-on-a-chip lined by human intestinal epithelial cells that experiences peristalsis-like motions and trickling fluid flow. Also, we discuss experimental techniques that can be used to analyze the cells in these organ-on-chip devices.
Collapse
|
122
|
van der Meer AD, Orlova VV, ten Dijke P, van den Berg A, Mummery CL. Three-dimensional co-cultures of human endothelial cells and embryonic stem cell-derived pericytes inside a microfluidic device. LAB ON A CHIP 2013; 13:3562-8. [PMID: 23702711 DOI: 10.1039/c3lc50435b] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Organs-on-chips are microengineered in vitro tissue structures that can be used as platforms for physiological and pathological research. They provide tissue-like microenvironments in which different cell types can be co-cultured in a controlled manner to create synthetic organ mimics. Blood vessels are an integral part of all tissues in the human body. Development of vascular structures is therefore an important research topic for advancing the field of organs-on-chips since generated tissues will require a blood or nutrient supply. Here, we have engineered three-dimensional constructs of vascular tissue inside microchannels by injecting a mixture of human umbilical vein endothelial cells, human embryonic stem cell-derived pericytes (the precursors of vascular smooth muscle cells) and rat tail collagen I into a polydimethylsiloxane microfluidic channel with dimensions 500 μm × 120 μm × 1 cm (w × h × l). Over the course of 12 h, the cells organized themselves into a single long tube resembling a blood vessel that followed the contours of the channel. Detailed examination of tube morphology by confocal microscopy revealed a mature endothelial monolayer with complete PECAM-1 staining at cell-cell contacts and pericytes incorporated inside the tubular structures. We also demonstrated that tube formation was disrupted in the presence of a neutralizing antibody against transforming growth factor-beta (TGF-β). The TGF-β signaling pathway is essential for normal vascular development; deletion of any of its components in mouse development results in defective vasculogenesis and angiogenesis and mutations in humans have been linked to multiple vascular genetic diseases. In the engineered microvessels, inhibition of TGF-β signaling resulted in tubes with smaller diameters and higher tortuosity, highly reminiscent of the abnormal vessels observed in patients with one particular vascular disease known as hereditary hemorrhagic telangiectasia (HHT). In summary, we have developed microengineered three-dimensional vascular structures that can be used as a model to test the effects of drugs and study the interaction between different human vascular cell types. In the future, the model may be integrated into larger tissue constructs to advance the development of organs-on-chips.
Collapse
Affiliation(s)
- Andries D van der Meer
- BIOS/Lab on a Chip, MESA+ Institute for Nanotechnology, University of Twente, The Netherlands.
| | | | | | | | | |
Collapse
|
123
|
Materne EM, Tonevitsky AG, Marx U. Chip-based liver equivalents for toxicity testing--organotypicalness versus cost-efficient high throughput. LAB ON A CHIP 2013; 13:3481-95. [PMID: 23722971 DOI: 10.1039/c3lc50240f] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Drug-induced liver toxicity dominates the reasons for pharmaceutical product ban, withdrawal or non-approval since the thalidomide disaster in the late-1950s. Hopes to finally solve the liver toxicity test dilemma have recently risen to a historic level based on the latest progress in human microfluidic tissue culture devices. Chip-based human liver equivalents are envisaged to identify liver toxic agents regularly undiscovered by current test procedures at industrial throughput. In this review, we focus on advanced microfluidic microscale liver equivalents, appraising them against the level of architectural and, consequently, functional identity with their human counterpart in vivo. We emphasise the inherent relationship between human liver architecture and its drug-induced injury. Furthermore, we plot the current socio-economic drug development environment against the possible value such systems may add. Finally, we try to sketch a forecast for translational innovations in the field.
Collapse
Affiliation(s)
- Eva-Maria Materne
- Technische Universität Berlin, Institute of Biotechnology, Department Medical Biotechnology, Gustav-Meyer-Allee 25, 13355 Berlin, Germany.
| | | | | |
Collapse
|
124
|
Harink B, Le Gac S, Truckenmüller R, van Blitterswijk C, Habibovic P. Regeneration-on-a-chip? The perspectives on use of microfluidics in regenerative medicine. LAB ON A CHIP 2013; 13:3512-28. [PMID: 23877890 DOI: 10.1039/c3lc50293g] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The aim of regenerative medicine is to restore or establish normal function of damaged tissues or organs. Tremendous efforts are placed into development of novel regenerative strategies, involving (stem) cells, soluble factors, biomaterials or combinations thereof, as a result of the growing need caused by continuous population aging. To satisfy this need, fast and reliable assessment of (biological) performance is sought, not only to select the potentially interesting candidates, but also to rule out poor ones at an early stage of development. Microfluidics may provide a new avenue to accelerate research and development in the field of regenerative medicine as it has proven its maturity for the realization of high-throughput screening platforms. In addition, microfluidic systems offer other advantages such as the possibility to create in vivo-like microenvironments. Besides the complexity of organs or tissues that need to be regenerated, regenerative medicine brings additional challenges of complex regeneration processes and strategies. The question therefore arises whether so much complexity can be integrated into microfluidic systems without compromising reliability and throughput of assays. With this review, we aim to investigate whether microfluidics can become widely applied in regenerative medicine research and/or strategies.
Collapse
Affiliation(s)
- Björn Harink
- Department of Tissue Regeneration, MIRA Institute for Biomedical Engineering and Technical Medicine, PO Box 217, 7500AE Enschede, The Netherlands.
| | | | | | | | | |
Collapse
|
125
|
Agarwal A, Goss JA, Cho A, McCain ML, Parker KK. Microfluidic heart on a chip for higher throughput pharmacological studies. LAB ON A CHIP 2013; 13:3599-608. [PMID: 23807141 PMCID: PMC3786400 DOI: 10.1039/c3lc50350j] [Citation(s) in RCA: 338] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We present the design of a higher throughput "heart on a chip" which utilizes a semi-automated fabrication technique to process sub millimeter sized thin film cantilevers of soft elastomers. Anisotropic cardiac microtissues which recapitulate the laminar architecture of the heart ventricle are engineered on these cantilevers. Deflection of these cantilevers, termed Muscular Thin Films (MTFs), during muscle contraction allows calculation of diastolic and systolic stresses generated by the engineered tissues. We also present the design of a reusable one channel fluidic microdevice completely built out of autoclavable materials which incorporates various features required for an optical cardiac contractility assay: metallic base which fits on a heating element for temperature control, transparent top for recording cantilever deformation and embedded electrodes for electrical field stimulation of the tissue. We employ the microdevice to test the positive inotropic effect of isoproterenol on cardiac contractility at dosages ranging from 1 nM to 100 μM. The higher throughput fluidic heart on a chip has applications in testing of cardiac tissues built from rare or expensive cell sources and for integration with other organ mimics. These advances will help alleviate translational barriers for commercial adoption of these technologies by improving the throughput and reproducibility of readout, standardization of the platform and scalability of manufacture.
Collapse
Affiliation(s)
- Ashutosh Agarwal
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Josue Adrian Goss
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Alexander Cho
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Megan Laura McCain
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute of Biologically Inspired Engineering, Harvard Stem Cell Institute, School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall Rm 321, Cambridge, MA, 02138, USA
- ; Fax: +(617) 495-9837; Tel: +(617) 495-2850
| |
Collapse
|
126
|
Lee JB, Sung JH. Organ-on-a-chip technology and microfluidic whole-body models for pharmacokinetic drug toxicity screening. Biotechnol J 2013; 8:1258-66. [PMID: 24038956 DOI: 10.1002/biot.201300086] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/30/2013] [Accepted: 07/07/2013] [Indexed: 01/19/2023]
Abstract
Microscale cell culture platforms better mimic the in vivo cellular microenvironment than conventional, macroscale systems. Microscale cultures therefore elicit a more authentic response from cultured cells, enabling physiologically realistic in vitro tissue models to be constructed. The fabrication of interconnecting microchambers and microchannels allows drug absorption, distribution, metabolism and elimination to be simulated, and enables precise manipulation of fluid flow to replicate blood circulation. Complex, multi-organ interactions can be investigated using "organ-on-a-chip" toxicology screens. By reproducing the dynamics of multi-organ interaction, the dynamics of various diseases and drug activities can be studied in mechanistic detail. In this review, we summarize the current status of technologies related to pharmacokinetic-based drug toxicity testing, and the use of microtechnology for reproducing the interaction between multiple organs.
Collapse
Affiliation(s)
- Jong Bum Lee
- University of Seoul, Chemical Engineering, Seoul, Korea
| | | |
Collapse
|
127
|
Vrana NE, Lavalle P, Dokmeci MR, Dehghani F, Ghaemmaghami AM, Khademhosseini A. Engineering functional epithelium for regenerative medicine and in vitro organ models: a review. TISSUE ENGINEERING PART B-REVIEWS 2013; 19:529-43. [PMID: 23705900 DOI: 10.1089/ten.teb.2012.0603] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent advances in the fields of microfabrication, biomaterials, and tissue engineering have provided new opportunities for developing biomimetic and functional tissues with potential applications in disease modeling, drug discovery, and replacing damaged tissues. An intact epithelium plays an indispensable role in the functionality of several organs such as the trachea, esophagus, and cornea. Furthermore, the integrity of the epithelial barrier and its degree of differentiation would define the level of success in tissue engineering of other organs such as the bladder and the skin. In this review, we focus on the challenges and requirements associated with engineering of epithelial layers in different tissues. Functional epithelial layers can be achieved by methods such as cell sheets, cell homing, and in situ epithelialization. However, for organs composed of several tissues, other important factors such as (1) in vivo epithelial cell migration, (2) multicell-type differentiation within the epithelium, and (3) epithelial cell interactions with the underlying mesenchymal cells should also be considered. Recent successful clinical trials in tissue engineering of the trachea have highlighted the importance of a functional epithelium for long-term success and survival of tissue replacements. Hence, using the trachea as a model tissue in clinical use, we describe the optimal structure of an artificial epithelium as well as challenges of obtaining a fully functional epithelium in macroscale. One of the possible remedies to address such challenges is the use of bottom-up fabrication methods to obtain a functional epithelium. Modular approaches for the generation of functional epithelial layers are reviewed and other emerging applications of microscale epithelial tissue models for studying epithelial/mesenchymal interactions in healthy and diseased (e.g., cancer) tissues are described. These models can elucidate the epithelial/mesenchymal tissue interactions at the microscale and provide the necessary tools for the next generation of multicellular engineered tissues and organ-on-a-chip systems.
Collapse
Affiliation(s)
- Nihal E Vrana
- 1 Institut National de la Santé et de la Recherche Médicale , INSERM, UMR-S 1121, "Biomatériaux et Bioingénierie," Strasbourg Cedex, France
| | | | | | | | | | | |
Collapse
|
128
|
Abstract
This review surveys selected methods of manufacture and applications of microdevices-miniaturized functional devices capable of handling cell and tissue cultures or producing particles-and discusses their potential relevance to nanomedicine. Many characteristics of microdevices such as miniaturization, increased throughput, and the ability to mimic organ-specific microenvironments are promising for the rapid, low-cost evaluation of the efficacy and toxicity of nanomaterials. Their potential to accurately reproduce the physiological environments that occur in vivo could reduce dependence on animal models in pharmacological testing. Technologies in microfabrications and microfluidics are widely applicable for nanomaterial synthesis and for the development of diagnostic devices. Although the use of microdevices in nanomedicine is still in its infancy, these technologies show promise for enhancing fundamental and applied research in nanomedicine.
Collapse
Affiliation(s)
- Michinao Hashimoto
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
129
|
Sung JH, Esch MB, Prot JM, Long CJ, Smith A, Hickman JJ, Shuler ML. Microfabricated mammalian organ systems and their integration into models of whole animals and humans. LAB ON A CHIP 2013; 13:1201-12. [PMID: 23388858 PMCID: PMC3593746 DOI: 10.1039/c3lc41017j] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
While in vitro cell based systems have been an invaluable tool in biology, they often suffer from a lack of physiological relevance. The discrepancy between the in vitro and in vivo systems has been a bottleneck in drug development process and biological sciences. The recent progress in microtechnology has enabled manipulation of cellular environment at a physiologically relevant length scale, which has led to the development of novel in vitro organ systems, often termed 'organ-on-a-chip' systems. By mimicking the cellular environment of in vivo tissues, various organ-on-a-chip systems have been reported to reproduce target organ functions better than conventional in vitro model systems. Ultimately, these organ-on-a-chip systems will converge into multi-organ 'body-on-a-chip' systems composed of functional tissues that reproduce the dynamics of the whole-body response. Such microscale in vitro systems will open up new possibilities in medical science and in the pharmaceutical industry.
Collapse
Affiliation(s)
- Jong H Sung
- Chemical Engineering, Hongik University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
130
|
Lee J, Kim SH, Kim YC, Choi I, Sung JH. Fabrication and characterization of microfluidic liver-on-a-chip using microsomal enzymes. Enzyme Microb Technol 2013; 53:159-64. [PMID: 23830456 DOI: 10.1016/j.enzmictec.2013.02.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/24/2013] [Accepted: 02/25/2013] [Indexed: 11/27/2022]
Abstract
Biotransformation in the liver plays an important role in determining the pharmacokinetic profile of drugs and food components. Current in vitro platforms for testing the liver metabolism suffers from the lack of resemblance to the human liver metabolism, mainly due to the lost metabolic activity of cultured hepatocytes and the absence of transport phenomena that occurs in the liver tissue. Here we report a microfluidic device with liver microsome encapsulated in 3-D hydrogel matrix, which can mimic the metabolism reaction and the transport phenomena in the liver. Photopolymerization of poly(ethylene glycol) diacrylate (PEG-DA) allows controlling the mass transfer with matrix sizes, and a gravity-induced passive flow can reproduce the blood flow through the liver. We measured the reaction kinetics of P450 enzymes in the device, and simulated the convection-diffusion-reaction characteristics inside the device with a mathematical model. Combination of mathematical analytical tool and the experimental tool allowed us to analyze and optimize the reaction kinetics inside the microfluidic chip. This novel in vitro platform can serve as a tool for screening the liver metabolism of various compounds.
Collapse
Affiliation(s)
- Jungwoo Lee
- I608, Department of Chemical Engineering, Hongik University, Sangsu-dong, Mapo-gu, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
131
|
Wikswo JP, Block FE, Cliffel DE, Goodwin CR, Marasco CC, Markov DA, McLean DL, McLean JA, McKenzie JR, Reiserer RS, Samson PC, Schaffer DK, Seale KT, Sherrod SD. Engineering challenges for instrumenting and controlling integrated organ-on-chip systems. IEEE Trans Biomed Eng 2013; 60:682-90. [PMID: 23380852 PMCID: PMC3696887 DOI: 10.1109/tbme.2013.2244891] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The sophistication and success of recently reported microfabricated organs-on-chips and human organ constructs have made it possible to design scaled and interconnected organ systems that may significantly augment the current drug development pipeline and lead to advances in systems biology. Physiologically realistic live microHuman (μHu) and milliHuman (mHu) systems operating for weeks to months present exciting and important engineering challenges such as determining the appropriate size for each organ to ensure appropriate relative organ functional activity, achieving appropriate cell density, providing the requisite universal perfusion media, sensing the breadth of physiological responses, and maintaining stable control of the entire system, while maintaining fluid scaling that consists of ~5 mL for the mHu and ~5 μL for the μHu. We believe that successful mHu and μHu systems for drug development and systems biology will require low-volume microdevices that support chemical signaling, microfabricated pumps, valves and microformulators, automated optical microscopy, electrochemical sensors for rapid metabolic assessment, ion mobility-mass spectrometry for real-time molecular analysis, advanced bioinformatics, and machine learning algorithms for automated model inference and integrated electronic control. Toward this goal, we are building functional prototype components and are working toward top-down system integration.
Collapse
Affiliation(s)
- John P. Wikswo
- Departments of Biomedical Engineering, Molecular Physiology & Biophysics, and Physics, and Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Frank E. Block
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - David E. Cliffel
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | - Cody R. Goodwin
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | - Christina C. Marasco
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - Dmitry A. Markov
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232-6840 USA
| | - David L. McLean
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235-1822 USA
| | | | - Ronald S. Reiserer
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Philip C. Samson
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - David K. Schaffer
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| | - Kevin T. Seale
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235-1631 USA
| | - Stacy D. Sherrod
- Department of Physics & Astronomy, Vanderbilt University, Nashville, TN 37235-1807 USA
| |
Collapse
|
132
|
Sivagnanam V, Gijs MAM. Exploring Living Multicellular Organisms, Organs, and Tissues Using Microfluidic Systems. Chem Rev 2013; 113:3214-47. [DOI: 10.1021/cr200432q] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Martin A. M. Gijs
- Laboratory
of Microsystems, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne,
Switzerland
| |
Collapse
|
133
|
|
134
|
Au SH, Fobel R, Desai SP, Voldman J, Wheeler AR. Cellular bias on the microscale: probing the effects of digital microfluidic actuation on mammalian cell health, fitness and phenotype. Integr Biol (Camb) 2013; 5:1014-25. [DOI: 10.1039/c3ib40104a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
135
|
|
136
|
Kumar Mahto S, Tenenbaum-Katan J, Sznitman J. Respiratory physiology on a chip. SCIENTIFICA 2012; 2012:364054. [PMID: 24278686 PMCID: PMC3820443 DOI: 10.6064/2012/364054] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/21/2012] [Indexed: 05/12/2023]
Abstract
Our current understanding of respiratory physiology and pathophysiological mechanisms of lung diseases is often limited by challenges in developing in vitro models faithful to the respiratory environment, both in cellular structure and physiological function. The recent establishment and adaptation of microfluidic-based in vitro devices (μFIVDs) of lung airways have enabled a wide range of developments in modern respiratory physiology. In this paper, we address recent efforts over the past decade aimed at advancing in vitro models of lung structure and airways using microfluidic technology and discuss their applications. We specifically focus on μFIVDs covering four major areas of respiratory physiology, namely, artificial lungs (AL), the air-liquid interface (ALI), liquid plugs and cellular injury, and the alveolar-capillary barrier (ACB).
Collapse
Affiliation(s)
- Sanjeev Kumar Mahto
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Janna Tenenbaum-Katan
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Josué Sznitman
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
137
|
Microfabrication technologies for oral drug delivery. Adv Drug Deliv Rev 2012; 64:496-507. [PMID: 22166590 DOI: 10.1016/j.addr.2011.11.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/18/2011] [Accepted: 11/28/2011] [Indexed: 12/21/2022]
Abstract
Micro-/nanoscale technologies such as lithographic techniques and microfluidics offer promising avenues to revolutionalize the fields of tissue engineering, drug discovery, diagnostics and personalized medicine. Microfabrication techniques are being explored for drug delivery applications due to their ability to combine several features such as precise shape and size into a single drug delivery vehicle. They also offer to create unique asymmetrical features incorporated into single or multiple reservoir systems maximizing contact area with the intestinal lining. Combined with intelligent materials, such microfabricated platforms can be designed to be bioadhesive and stimuli-responsive. Apart from drug delivery devices, microfabrication technologies offer exciting opportunities to create biomimetic gastrointestinal tract models incorporating physiological cell types, flow patterns and brush-border like structures. Here we review the recent developments in this field with a focus on the applications of microfabrication in the development of oral drug delivery devices and biomimetic gastrointestinal tract models that can be used to evaluate the drug delivery efficacy.
Collapse
|