1
|
Esfahani SN, Resto Irizarry AM, Xue X, Lee SBD, Shao Y, Fu J. Micro/nanoengineered technologies for human pluripotent stem cells maintenance and differentiation. NANO TODAY 2021; 41:101310. [PMID: 34745321 PMCID: PMC8570530 DOI: 10.1016/j.nantod.2021.101310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Human pluripotent stem cells (hPSCs) are a promising source of cells for cell replacement-based therapies as well as modeling human development and diseases in vitro. However, achieving fate control of hPSC with a high yield and specificity remains challenging. The fate specification of hPSCs is regulated by biochemical and biomechanical cues in their environment. Driven by this knowledge, recent exciting advances in micro/nanoengineering have been leveraged to develop a broad range of tools for the generation of extracellular biomechanical and biochemical signals that determine the behavior of hPSCs. In this review, we summarize such micro/nanoengineered technologies for controlling hPSC fate and highlight the role of biochemical and biomechanical cues such as substrate rigidity, surface topography, and cellular confinement in the hPSC-based technologies that are on the horizon.
Collapse
Affiliation(s)
- Sajedeh Nasr Esfahani
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Samuel Byung-Deuk Lee
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yue Shao
- Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Jiangping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
2
|
Shakeri A, Khan S, Didar TF. Conventional and emerging strategies for the fabrication and functionalization of PDMS-based microfluidic devices. LAB ON A CHIP 2021; 21:3053-3075. [PMID: 34286800 DOI: 10.1039/d1lc00288k] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Microfluidics is an emerging and multidisciplinary field that is of great interest to manufacturers in medicine, biotechnology, and chemistry, as it provides unique tools for the development of point-of-care diagnostics, organs-on-chip systems, and biosensors. Polymeric microfluidics, unlike glass and silicon, offer several advantages such as low-cost mass manufacturing and a wide range of beneficial material properties, which make them the material of choice for commercial applications and high-throughput systems. Among polymers used for the fabrication of microfluidic devices, polydimethylsiloxane (PDMS) still remains the most widely used material in academia due to its advantageous properties, such as excellent transparency and biocompatibility. However, commercialization of PDMS has been a challenge mostly due to the high cost of the current fabrication strategies. Moreover, specific surface modification and functionalization steps are required to tailor the surface chemistry of PDMS channels (e.g. biomolecule immobilization, surface hydrophobicity and antifouling properties) with respect to the desired application. While significant research has been reported in the field of PDMS microfluidics, functionalization of PDMS surfaces remains a critical step in the fabrication process that is difficult to navigate. This review first offers a thorough illustration of existing fabrication methods for PDMS-based microfluidic devices, providing several recent advancements in this field with the aim of reducing the cost and time for mass production of these devices. Next, various conventional and emerging approaches for engineering the surface chemistry of PDMS are discussed in detail. We provide a wide range of functionalization techniques rendering PDMS microchannels highly biocompatible for physical or covalent immobilization of various biological entities while preventing non-specific interactions.
Collapse
Affiliation(s)
- Amid Shakeri
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
| | - Shadman Khan
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Tohid F Didar
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada.
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
3
|
Aranda Hernandez J, Heuer C, Bahnemann J, Szita N. Microfluidic Devices as Process Development Tools for Cellular Therapy Manufacturing. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 179:101-127. [PMID: 34410457 DOI: 10.1007/10_2021_169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cellular therapies are creating a paradigm shift in the biomanufacturing industry. Particularly for autologous therapies, small-scale processing methods are better suited than the large-scale approaches that are traditionally employed in the industry. Current small-scale methods for manufacturing personalized cell therapies, however, are labour-intensive and involve a number of 'open events'. To overcome these challenges, new cell manufacturing platforms following a GMP-in-a-box concept have recently come on the market (GMP: Good Manufacturing Practice). These are closed automated systems with built-in pumps for fluid handling and sensors for in-process monitoring. At a much smaller scale, microfluidic devices exhibit many of the same features as current GMP-in-a-box systems. They are closed systems, fluids can be processed and manipulated, and sensors integrated for real-time detection of process variables. Fabricated from polymers, they can be made disposable, i.e. single-use. Furthermore, microfluidics offers exquisite spatiotemporal control over the cellular microenvironment, promising both reproducibility and control of outcomes. In this chapter, we consider the challenges in cell manufacturing, highlight recent advances of microfluidic devices for each of the main process steps, and summarize our findings on the current state of the art. As microfluidic cell culture devices have been reported for both adherent and suspension cell cultures, we report on devices for the key process steps, or unit operations, of both stem cell therapies and cell-based immunotherapies.
Collapse
Affiliation(s)
| | - Christopher Heuer
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Janina Bahnemann
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Nicolas Szita
- Biochemical Engineering Department, University College London (UCL), London, UK.
| |
Collapse
|
4
|
Cui KW, Engel L, Dundes CE, Nguyen TC, Loh KM, Dunn AR. Spatially controlled stem cell differentiation via morphogen gradients: A comparison of static and dynamic microfluidic platforms. JOURNAL OF VACUUM SCIENCE & TECHNOLOGY. A, VACUUM, SURFACES, AND FILMS : AN OFFICIAL JOURNAL OF THE AMERICAN VACUUM SOCIETY 2020; 38:033205. [PMID: 32255900 PMCID: PMC7093209 DOI: 10.1116/1.5142012#suppl] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 05/30/2023]
Abstract
The ability to harness the processes by which complex tissues arise during embryonic development would improve the ability to engineer complex tissuelike constructs in vitro-a longstanding goal of tissue engineering and regenerative medicine. In embryos, uniform populations of stem cells are exposed to spatial gradients of diffusible extracellular signaling proteins, known as morphogens. Varying levels of these signaling proteins induce stem cells to differentiate into distinct cell types at different positions along the gradient, thus creating spatially patterned tissues. Here, the authors describe two straightforward and easy-to-adopt microfluidic strategies to expose human pluripotent stem cells in vitro to spatial gradients of desired differentiation-inducing extracellular signals. Both approaches afford a high degree of control over the distribution of extracellular signals, while preserving the viability of the cultured stem cells. The first microfluidic platform is commercially available and entails static culture, whereas the second microfluidic platform requires fabrication and dynamic fluid exchange. In each platform, the authors first computationally modeled the spatial distribution of differentiation-inducing extracellular signals. Then, the authors used each platform to expose human pluripotent stem cells to a gradient of these signals (in this case, inducing a cell type known as the primitive streak), resulting in a regionalized culture with differentiated primitive streak cells predominately localized on one side and undifferentiated stem cells at the other side of the device. By combining this approach with a fluorescent reporter for differentiated cells and live-cell fluorescence imaging, the authors characterized the spatial and temporal dynamics of primitive streak differentiation within the induced signaling gradients. Microfluidic approaches to create precisely controlled morphogen gradients will add to the stem cell and developmental biology toolkit, and may eventually pave the way to create increasingly spatially patterned tissuelike constructs in vitro.
Collapse
Affiliation(s)
- Kiara W Cui
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Leeya Engel
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Carolyn E Dundes
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305
| | - Tina C Nguyen
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Kyle M Loh
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| |
Collapse
|
5
|
Cui KW, Engel L, Dundes CE, Nguyen TC, Loh KM, Dunn AR. Spatially controlled stem cell differentiation via morphogen gradients: A comparison of static and dynamic microfluidic platforms. JOURNAL OF VACUUM SCIENCE & TECHNOLOGY. A, VACUUM, SURFACES, AND FILMS : AN OFFICIAL JOURNAL OF THE AMERICAN VACUUM SOCIETY 2020; 38:033205. [PMID: 32255900 PMCID: PMC7093209 DOI: 10.1116/1.5142012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 05/21/2023]
Abstract
The ability to harness the processes by which complex tissues arise during embryonic development would improve the ability to engineer complex tissuelike constructs in vitro-a longstanding goal of tissue engineering and regenerative medicine. In embryos, uniform populations of stem cells are exposed to spatial gradients of diffusible extracellular signaling proteins, known as morphogens. Varying levels of these signaling proteins induce stem cells to differentiate into distinct cell types at different positions along the gradient, thus creating spatially patterned tissues. Here, the authors describe two straightforward and easy-to-adopt microfluidic strategies to expose human pluripotent stem cells in vitro to spatial gradients of desired differentiation-inducing extracellular signals. Both approaches afford a high degree of control over the distribution of extracellular signals, while preserving the viability of the cultured stem cells. The first microfluidic platform is commercially available and entails static culture, whereas the second microfluidic platform requires fabrication and dynamic fluid exchange. In each platform, the authors first computationally modeled the spatial distribution of differentiation-inducing extracellular signals. Then, the authors used each platform to expose human pluripotent stem cells to a gradient of these signals (in this case, inducing a cell type known as the primitive streak), resulting in a regionalized culture with differentiated primitive streak cells predominately localized on one side and undifferentiated stem cells at the other side of the device. By combining this approach with a fluorescent reporter for differentiated cells and live-cell fluorescence imaging, the authors characterized the spatial and temporal dynamics of primitive streak differentiation within the induced signaling gradients. Microfluidic approaches to create precisely controlled morphogen gradients will add to the stem cell and developmental biology toolkit, and may eventually pave the way to create increasingly spatially patterned tissuelike constructs in vitro.
Collapse
Affiliation(s)
- Kiara W Cui
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Leeya Engel
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Carolyn E Dundes
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305
| | - Tina C Nguyen
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| | - Kyle M Loh
- Department of Developmental Biology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California 94305
| |
Collapse
|
6
|
Kim MH, Kino-Oka M. Bioengineering Considerations for a Nurturing Way to Enhance Scalable Expansion of Human Pluripotent Stem Cells. Biotechnol J 2020; 15:e1900314. [PMID: 31904180 DOI: 10.1002/biot.201900314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Understanding how defects in mechanotransduction affect cell-to-cell variability will add to the fundamental knowledge of human pluripotent stem cell (hPSC) culture, and may suggest new approaches for achieving a robust, reproducible, and scalable process that result in consistent product quality and yields. Here, the current state of the understanding of the fundamental mechanisms that govern the growth kinetics of hPSCs between static and dynamic cultures is reviewed, the factors causing fluctuations are identified, and culture strategies that might eliminate or minimize the occurrence of cell-to-cell variability arising from these fluctuations are discussed. The existing challenges in the development of hPSC expansion methods for enabling the transition from process development to large-scale production are addressed, a mandatory step for industrial and clinical applications of hPSCs.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
7
|
An S, Han SY, Cho SW. Hydrogel-integrated Microfluidic Systems for Advanced Stem Cell Engineering. BIOCHIP JOURNAL 2019. [DOI: 10.1007/s13206-019-3402-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
8
|
Liu W, Deng C, Godoy-Parejo C, Zhang Y, Chen G. Developments in cell culture systems for human pluripotent stem cells. World J Stem Cells 2019; 11:968-981. [PMID: 31768223 PMCID: PMC6851012 DOI: 10.4252/wjsc.v11.i11.968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) are important resources for cell-based therapies and pharmaceutical applications. In order to realize the potential of hPSCs, it is critical to develop suitable technologies required for specific applications. Most hPSC technologies depend on cell culture, and are critically influenced by culture medium composition, extracellular matrices, handling methods, and culture platforms. This review summarizes the major technological advances in hPSC culture, and highlights the opportunities and challenges in future therapeutic applications.
Collapse
Affiliation(s)
- Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Chunhao Deng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Carlos Godoy-Parejo
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau 999078, China.
| |
Collapse
|
9
|
Marx U, Walles H, Hoffmann S, Lindner G, Horland R, Sonntag F, Klotzbach U, Sakharov D, Tonevitsky A, Lauster R. ‘Human-on-a-chip’ Developments: A Translational Cutting-edge Alternative to Systemic Safety Assessment and Efficiency Evaluation of Substances in Laboratory Animals and Man? Altern Lab Anim 2019; 40:235-57. [DOI: 10.1177/026119291204000504] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Uwe Marx
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | | | - Silke Hoffmann
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Gerd Lindner
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Reyk Horland
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| | - Frank Sonntag
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | - Udo Klotzbach
- Fraunhofer-Institut für Werkstoff- und Strahltechnik IWS, Dresden, Germany
| | | | | | - Roland Lauster
- Technische Universität Berlin, Department of Biotechnology, Berlin, Germany
| |
Collapse
|
10
|
Varma S, Voldman J. Caring for cells in microsystems: principles and practices of cell-safe device design and operation. LAB ON A CHIP 2018; 18:3333-3352. [PMID: 30324208 PMCID: PMC6254237 DOI: 10.1039/c8lc00746b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Microfluidic device designers and users continually question whether cells are 'happy' in a given microsystem or whether they are perturbed by micro-scale technologies. This issue is normally brought up by engineers building platforms, or by external reviewers (academic or commercial) comparing multiple technological approaches to a problem. Microsystems can apply combinations of biophysical and biochemical stimuli that, although essential to device operation, may damage cells in complex ways. However, assays to assess the impact of microsystems upon cells have been challenging to conduct and have led to subjective interpretation and evaluation of cell stressors, hampering development and adoption of microsystems. To this end, we introduce a framework that defines cell health, describes how device stimuli may stress cells, and contrasts approaches to measure cell stress. Importantly, we provide practical guidelines regarding device design and operation to minimize cell stress, and recommend a minimal set of quantitative assays that will enable standardization in the assessment of cell health in diverse devices. We anticipate that as microsystem designers, reviewers, and end-users enforce such guidelines, we as a community can create a set of essential principles that will further the adoption of such technologies in clinical, translational and commercial applications.
Collapse
Affiliation(s)
- Sarvesh Varma
- Department of Electrical Engineering and Computer Science
, Massachusetts Institute of Technology
,
77 Massachusetts Avenue, Room 36-824
, Cambridge
, USA
.
; Fax: +617 258 5846
; Tel: +617 253 1583
| | - Joel Voldman
- Department of Electrical Engineering and Computer Science
, Massachusetts Institute of Technology
,
77 Massachusetts Avenue, Room 36-824
, Cambridge
, USA
.
; Fax: +617 258 5846
; Tel: +617 253 1583
| |
Collapse
|
11
|
Chen KG, Mallon BS, Park K, Robey PG, McKay RDG, Gottesman MM, Zheng W. Pluripotent Stem Cell Platforms for Drug Discovery. Trends Mol Med 2018; 24:805-820. [PMID: 30006147 DOI: 10.1016/j.molmed.2018.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/30/2022]
Abstract
Use of human pluripotent stem cells (hPSCs) and their differentiated derivatives have led to recent proof-of-principle drug discoveries, defining a pathway to the implementation of hPSC-based drug discovery (hPDD). Current hPDD strategies, however, have inevitable conceptual biases and technological limitations, including the dimensionality of cell-culture methods, cell maturity and functionality, experimental variability, and data reproducibility. In this review, we dissect representative hPDD systems via analysis of hPSC-based 2D-monolayers, 3D culture, and organoids. We discuss mechanisms of drug discovery and drug repurposing, and roles of membrane drug transporters in tissue maturation and hPDD using the example of drugs that target various mutations of CFTR, the cystic fibrosis transmembrane conductance regulator gene, in patients with cystic fibrosis.
Collapse
Affiliation(s)
- Kevin G Chen
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara S Mallon
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyeyoon Park
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald D G McKay
- The Lieber Institute for Brain Development, Baltimore, MD 21205, USA
| | - Michael M Gottesman
- The Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|
12
|
Rico-Varela J, Ho D, Wan LQ. In Vitro Microscale Models for Embryogenesis. ADVANCED BIOSYSTEMS 2018; 2:1700235. [PMID: 30533517 PMCID: PMC6286056 DOI: 10.1002/adbi.201700235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 12/15/2022]
Abstract
Embryogenesis is a highly regulated developmental process requiring complex mechanical and biochemical microenvironments to give rise to a fully developed and functional embryo. Significant efforts have been taken to recapitulate specific features of embryogenesis by presenting the cells with developmentally relevant signals. The outcomes, however, are limited partly due to the complexity of this biological process. Microtechnologies such as micropatterned and microfluidic systems, along with new emerging embryonic stem cell-based models, could potentially serve as powerful tools to study embryogenesis. The aim of this article is to review major studies involving the culturing of pluripotent stem cells using different geometrical patterns, microfluidic platforms, and embryo/embryoid body-on-a-chip modalities. Indeed, new research opportunities have emerged for establishing in vitro culture for studying human embryogenesis and for high-throughput pharmacological testing platforms and disease models to prevent defects in early stages of human development.
Collapse
Affiliation(s)
- Jennifer Rico-Varela
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Dominic Ho
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| | - Leo Q. Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
- Center for Modeling, Simulation and Imaging in Medicine, Rensselaer Polytechnic Institute, 110 8th Street, Troy NY 12180
| |
Collapse
|
13
|
Osaki T, Shin Y, Sivathanu V, Campisi M, Kamm RD. In Vitro Microfluidic Models for Neurodegenerative Disorders. Adv Healthc Mater 2018; 7. [PMID: 28881425 DOI: 10.1002/adhm.201700489] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/18/2017] [Indexed: 01/09/2023]
Abstract
Microfluidic devices enable novel means of emulating neurodegenerative disease pathophysiology in vitro. These organ-on-a-chip systems can potentially reduce animal testing and substitute (or augment) simple 2D culture systems. Reconstituting critical features of neurodegenerative diseases in a biomimetic system using microfluidics can thereby accelerate drug discovery and improve our understanding of the mechanisms of several currently incurable diseases. This review describes latest advances in modeling neurodegenerative diseases in the central nervous system and the peripheral nervous system. First, this study summarizes fundamental advantages of microfluidic devices in the creation of compartmentalized cell culture microenvironments for the co-culture of neurons, glial cells, endothelial cells, and skeletal muscle cells and in their recapitulation of spatiotemporal chemical gradients and mechanical microenvironments. Then, this reviews neurodegenerative-disease-on-a-chip models focusing on Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Finally, this study discusses about current drawbacks of these models and strategies that may overcome them. These organ-on-chip technologies can be useful to be the first line of testing line in drug development and toxicology studies, which can contribute significantly to minimize the phase of animal testing steps.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Yoojin Shin
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Vivek Sivathanu
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| | - Marco Campisi
- Department of Mechanical and Aerospace EngineeringPolitecnico di Torino Corso Duca degli Abruzzi 24 10129 Torino Italy
| | - Roger D. Kamm
- Department of Mechanical EngineeringMassachusetts Institutes of Technology 500 Technology Square MIT Building, Room NE47‐321 Cambridge MA 02139 USA
- Department of Biological EngineeringMassachusetts Institutes of Technology 500 Technology Square, MIT Building, Room NE47‐321 Cambridge MA 02139 USA
| |
Collapse
|
14
|
Advances in Micro- and Nanotechnologies for Stem Cell-Based Translational Applications. STEM CELL BIOLOGY AND REGENERATIVE MEDICINE 2017. [DOI: 10.1007/978-3-319-29149-9_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
|
16
|
Zhang Y, Wang X, Pong M, Chen L, Ye Z. Application of Bioreactor in Stem Cell Culture. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jbise.2017.1011037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
17
|
Kamei KI, Koyama Y, Tokunaga Y, Mashimo Y, Yoshioka M, Fockenberg C, Mosbergen R, Korn O, Wells C, Chen Y. Characterization of Phenotypic and Transcriptional Differences in Human Pluripotent Stem Cells under 2D and 3D Culture Conditions. Adv Healthc Mater 2016; 5:2951-2958. [PMID: 27775225 DOI: 10.1002/adhm.201600893] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 12/26/2022]
Abstract
Human pluripotent stem cells hold great promise for applications in drug discovery and regenerative medicine. Microfluidic technology is a promising approach for creating artificial microenvironments; however, although a proper 3D microenvironment is required to achieve robust control of cellular phenotypes, most current microfluidic devices provide only 2D cell culture and do not allow tuning of physical and chemical environmental cues simultaneously. Here, the authors report a 3D cellular microenvironment plate (3D-CEP), which consists of a microfluidic device filled with thermoresponsive poly(N-isopropylacrylamide)-β-poly(ethylene glycol) hydrogel (HG), which enables systematic tuning of both chemical and physical environmental cues as well as in situ cell monitoring. The authors show that H9 human embryonic stem cells (hESCs) and 253G1 human induced pluripotent stem cells in the HG/3D-CEP system maintain their pluripotent marker expression under HG/3D-CEP self-renewing conditions. Additionally, global gene expression analyses are used to elucidate small variations among different test environments. Interestingly, the authors find that treatment of H9 hESCs under HG/3D-CEP self-renewing conditions results in initiation of entry into the neural differentiation process by induction of PAX3 and OTX1 expression. The authors believe that this HG/3D-CEP system will serve as a versatile platform for developing targeted functional cell lines and facilitate advances in drug screening and regenerative medicine.
Collapse
Affiliation(s)
- Ken-ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Yoshie Koyama
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Yumie Tokunaga
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Yasumasa Mashimo
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Momoko Yoshioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Christopher Fockenberg
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
| | - Rowland Mosbergen
- Australia Institute for Biotechnology and Nanotechnology (AIBN); University of Queensland; Brisbane QLD 4072 Australia
- Department of Anatomy and Neuroscience; University of Melbourne; Melbourne Vic 3010 Australia
| | - Othmar Korn
- Australia Institute for Biotechnology and Nanotechnology (AIBN); University of Queensland; Brisbane QLD 4072 Australia
| | - Christine Wells
- Australia Institute for Biotechnology and Nanotechnology (AIBN); University of Queensland; Brisbane QLD 4072 Australia
- Department of Anatomy and Neuroscience; University of Melbourne; Melbourne Vic 3010 Australia
| | - Yong Chen
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS); Kyoto University; Kyoto 6068501 Japan
- Ecole Normale Supérieure; CNRS-ENS-UPMC UMR 8640; 24 Rue L'homond Paris 75005 France
| |
Collapse
|
18
|
Novel micro-photobioreactor design and monitoring method for assessing microalgae response to light intensity. ALGAL RES 2016. [DOI: 10.1016/j.algal.2016.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
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: 105] [Impact Index Per Article: 11.7] [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
|
20
|
Gagliano O, Elvassore N, Luni C. Microfluidic technology enhances the potential of human pluripotent stem cells. Biochem Biophys Res Commun 2016; 473:683-7. [DOI: 10.1016/j.bbrc.2015.12.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 12/15/2015] [Indexed: 01/02/2023]
|
21
|
Park D, Lim J, Park JY, Lee SH. Concise Review: Stem Cell Microenvironment on a Chip: Current Technologies for Tissue Engineering and Stem Cell Biology. Stem Cells Transl Med 2015; 4:1352-68. [PMID: 26450425 DOI: 10.5966/sctm.2015-0095] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 07/29/2015] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Stem cells have huge potential in many therapeutic areas. With conventional cell culture methods, however, it is difficult to achieve in vivo-like microenvironments in which a number of well-controlled stimuli are provided for growing highly sensitive stem cells. In contrast, microtechnology-based platforms offer advantages of high precision, controllability, scalability, and reproducibility, enabling imitation of the complex physiological context of in vivo. This capability may fill the gap between the present knowledge about stem cells and that required for clinical stem cell-based therapies. We reviewed the various types of microplatforms on which stem cell microenvironments are mimicked. We have assigned the various microplatforms to four categories based on their practical uses to assist stem cell biologists in using them for research. In particular, many examples are given of microplatforms used for the production of embryoid bodies and aggregates of stem cells in vitro. We also categorized microplatforms based on the types of factors controlling the behaviors of stem cells. Finally, we outline possible future directions for microplatform-based stem cell research, such as research leading to the production of well-defined environments for stem cells to be used in scaled-up systems or organs-on-a-chip, the regulation of induced pluripotent stem cells, and the study of the genetic states of stem cells on microplatforms. SIGNIFICANCE Stem cells are highly sensitive to a variety of physicochemical cues, and their fate can be easily altered by a slight change of environment; therefore, systematic analysis and discrimination of the extracellular signals and intracellular pathways controlling the fate of cells and experimental realization of sensitive and controllable niche environments are critical. This review introduces diverse microplatforms to provide in vitro stem cell niches. Microplatforms could control microenvironments around cells and have recently attracted much attention in biology including stem cell research. These microplatforms and the future directions of stem cell microenvironment are described.
Collapse
Affiliation(s)
- DoYeun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Jaeho Lim
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Joong Yull Park
- School of Mechanical Engineering, College of Engineering, Chung-ang University, Seoul, Republic of Korea
| | - Sang-Hoon Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea School of Biomedical Engineering, College of Health Science, Korea University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Qian T, Shusta EV, Palecek SP. Advances in microfluidic platforms for analyzing and regulating human pluripotent stem cells. Curr Opin Genet Dev 2015; 34:54-60. [PMID: 26313850 DOI: 10.1016/j.gde.2015.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 01/20/2023]
Abstract
Microfluidic devices employ submillimeter length scale control of flow to achieve high-resolution spatial and temporal control over the microenvironment, providing powerful tools to elucidate mechanisms of human pluripotent stem cell (hPSC) regulation and to elicit desired hPSC fates. In addition, microfluidics allow control of paracrine and juxtracrine signaling, thereby enabling fabrication of microphysiological systems comprised of multiple cell types organized into organs-on-a-chip. Microfluidic cell culture systems can also be integrated with actuators and sensors, permitting construction of high-density arrays of cell-based biosensors for screening applications. This review describes recent advances in using microfluidics to understand mechanisms by which the microenvironment regulates hPSC fates and applications of microfluidics to realize the potential of hPSCs for in vitro modeling and screening applications.
Collapse
Affiliation(s)
- Tongcheng Qian
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI 53706, USA.
| |
Collapse
|
23
|
Jastrzebska E, Tomecka E, Jesion I. Heart-on-a-chip based on stem cell biology. Biosens Bioelectron 2015; 75:67-81. [PMID: 26298640 DOI: 10.1016/j.bios.2015.08.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/28/2015] [Accepted: 08/08/2015] [Indexed: 12/26/2022]
Abstract
Heart diseases are one of the main causes of death around the world. The great challenge for scientists is to develop new therapeutic methods for these types of ailments. Stem cells (SCs) therapy could be one of a promising technique used for renewal of cardiac cells and treatment of heart diseases. Conventional in vitro techniques utilized for investigation of heart regeneration do not mimic natural cardiac physiology. Lab-on-a-chip systems may be the solution which could allow the creation of a heart muscle model, enabling the growth of cardiac cells in conditions similar to in vivo conditions. Microsystems can be also used for differentiation of stem cells into heart cells, successfully. It will help better understand of proliferation and regeneration ability of these cells. In this review, we present Heart-on-a-chip systems based on cardiac cell culture and stem cell biology. This review begins with the description of the physiological environment and the functions of the heart. Next, we shortly described conventional techniques of stem cells differentiation into the cardiac cells. This review is mostly focused on describing Lab-on-a-chip systems for cardiac tissue engineering. Therefore, in the next part of this article, the microsystems for both cardiac cell culture and SCs differentiation into cardiac cells are described. The section about SCs differentiation into the heart cells is divided in sections describing biochemical, physical and mechanical stimulations. Finally, we outline present challenges and future research concerning Heart-on-a-chip based on stem cell biology.
Collapse
Affiliation(s)
- Elzbieta Jastrzebska
- Institute of Biotechnology, Department of Microbioanalytics, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland.
| | - Ewelina Tomecka
- Institute of Biotechnology, Department of Microbioanalytics, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Iwona Jesion
- Department of Animal Environment Biology, Faculty of Animal Science, Warsaw University of Life Science, Ciszewskiego 8, 02-786 Warsaw, Poland
| |
Collapse
|
24
|
Sikorski DJ, Caron NJ, VanInsberghe M, Zahn H, Eaves CJ, Piret JM, Hansen CL. Clonal analysis of individual human embryonic stem cell differentiation patterns in microfluidic cultures. Biotechnol J 2015; 10:1546-54. [PMID: 26059045 DOI: 10.1002/biot.201500035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/04/2015] [Accepted: 06/05/2015] [Indexed: 01/23/2023]
Abstract
Heterogeneity in the clonal outputs of individual human embryonic stem cells (hESCs) confounds analysis of their properties in studies of bulk populations and how to manipulate them for clinical applications. To circumvent this problem we developed a microfluidic device that supports the robust generation of colonies derived from single ESCs. This microfluidic system contains 160 individually addressable chambers equipped for perfusion culture of individual hESCs that could be shown to match the growth rates, marker expression and colony morphologies obtained in conventional cultures. Use of this microfluidic device to analyze the clonal growth kinetics of multiple individual hESCs induced to differentiation revealed variable shifts in the growth rate, area per cell and expression of OCT4 in the progeny of individual hESCs. Interestingly, low OCT4 expression, a slower growth rate and low nuclear to cytoplasmic ratios were found to be correlated responses. This study demonstrates how microfluidic systems can be used to enable large scale live-cell imaging of isolated hESCs exposed to changing culture conditions, to examine how different aspects of their variable responses are correlated.
Collapse
Affiliation(s)
- Darek J Sikorski
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada.,Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Nicolas J Caron
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Michael VanInsberghe
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Hans Zahn
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - James M Piret
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Carl L Hansen
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada. .,Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
25
|
Giobbe GG, Michielin F, Luni C, Giulitti S, Martewicz S, Dupont S, Floreani A, Elvassore N. Functional differentiation of human pluripotent stem cells on a chip. Nat Methods 2015; 12:637-40. [DOI: 10.1038/nmeth.3411] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 03/10/2015] [Indexed: 12/20/2022]
|
26
|
Corti S, Faravelli I, Cardano M, Conti L. Human pluripotent stem cells as tools for neurodegenerative and neurodevelopmental disease modeling and drug discovery. Expert Opin Drug Discov 2015; 10:615-29. [PMID: 25891144 DOI: 10.1517/17460441.2015.1037737] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Although intensive efforts have been made, effective treatments for neurodegenerative and neurodevelopmental diseases have not been yet discovered. Possible reasons for this include the lack of appropriate disease models of human neurons and a limited understanding of the etiological and neurobiological mechanisms. Recent advances in pluripotent stem cell (PSC) research have now opened the path to the generation of induced pluripotent stem cells (iPSCs) starting from somatic cells, thus offering an unlimited source of patient-specific disease-relevant neuronal cells. AREAS COVERED In this review, the authors focus on the use of human PSC-derived cells in modeling neurological disorders and discovering of new drugs and provide their expert perspectives on the field. EXPERT OPINION The advent of human iPSC-based disease models has fuelled renewed enthusiasm and enormous expectations for insights of disease mechanisms and identification of more disease-relevant and novel molecular targets. Human PSCs offer a unique tool that is being profitably exploited for high-throughput screening (HTS) platforms. This process can lead to the identification and optimization of molecules/drugs and thus move forward new pharmacological therapies for a wide range of neurodegenerative and neurodevelopmental conditions. It is predicted that improvements in the production of mature neuronal subtypes, from patient-specific human-induced pluripotent stem cells and their adaptation to culture, to HTS platforms will allow the increased exploitation of human pluripotent stem cells in drug discovery programs.
Collapse
Affiliation(s)
- Stefania Corti
- University of Milan, Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation, Neurology Unit, IRCCS Foundation Ca'Granda Ospedale Maggiore Policlinico , via Francesco Sforza 35, Milan 20122 , Italy +39 02 55033817 ;
| | | | | | | |
Collapse
|
27
|
Yu DX, Marchetto MC, Gage FH. Therapeutic translation of iPSCs for treating neurological disease. Cell Stem Cell 2014; 12:678-88. [PMID: 23746977 DOI: 10.1016/j.stem.2013.05.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Somatic cellular reprogramming is a fast-paced and evolving field that is changing the way scientists approach neurological diseases. For the first time in the history of neuroscience, it is feasible to study the behavior of live neurons from patients with neurodegenerative diseases, such as Alzheimer's and Parkinson's disease, and neuropsychiatric diseases, such as autism and schizophrenia. In this Perspective, we will discuss reprogramming technology in the context of its potential use for modeling and treating neurological and psychiatric diseases and will highlight areas of caution and opportunities for improvement.
Collapse
Affiliation(s)
- Diana X Yu
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
28
|
Sun B, Yu W, Wang F, Song W, Jin H, Sun Y. Effects of group culture on the development of discarded human embryos and the construction of human embryonic stem cell lines. J Assist Reprod Genet 2014; 31:1369-76. [PMID: 25113620 DOI: 10.1007/s10815-014-0308-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/21/2014] [Indexed: 10/24/2022] Open
Abstract
PURPOSE To explore the effect of group culture on the developmental potential of discarded embryos in in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles and establish the human embryonic stem cell lines for future research. METHORDS Fresh discarded embryos were collected from the IVF/ICSI-ET program in the reproductive medical center of the first affiliated hospital of Zhengzhou university in this study. All zygotes were individually cultured from Day 1 to Day 3. On Day 3, discard embryos were then cultured in group of 1-4 embryos per droplet (30 μl/droplets) with a constant culture medium until Day 5 or 6. Mechanical method was used to isolate the inner cell mass (ICM) of blastocyst from the embryo. Then we inoculated the ICM on feeder layer. After identification of those cells, the human embryonic stem cell lines (hESCs) were established. RESULTS In this study, we collected 1,223 fresh discarded embryos and they were sequential cultured to the blastocysts (18.07 %, 221/1,223), in which good quality blastocysts were 61(4.98 %, 61/1,223). There was no significant difference in the patients. The embryos from 1PN, 2PN, 3PN were sequential cultured to the blastocyst s(39.31 %,92/234;12.87 %,64/497;13.21 %,65/492),in which good quality blastocysts was 13.6 %(32/92),2.61 %(13/64), 3.04 %(15/65).1PN embryo's blastulation rate and quality embryo formation rate was significantly higher than the 2PN and 3PN embryos' (P <0.05). Three embryos group cultivation has the highest blastulation rate and quality embryo formation rate (P <0.05). In total, we successfully established 4 hESCs lines. CONCLUSION The group culture of human discard embryos can improve the blastulation rate and blastocyst quality to some extent. Three embryos group cultivate is the better culture number. Human discard embryos are good source for establishment of hESCs.
Collapse
Affiliation(s)
- Bo Sun
- Reproductive Medical Center, First Affiliated Hospital of Zhengzhou University, Constructive East Road, Zhengzhou, 450052, China
| | | | | | | | | | | |
Collapse
|
29
|
Grygoryev K, Herzog G, Jackson N, Strutwolf J, Arrigan DWM, McDermott K, Galvin P. Reversible Integration of Microfluidic Devices with Microelectrode Arrays for Neurobiological Applications. BIONANOSCIENCE 2014. [DOI: 10.1007/s12668-014-0137-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
30
|
Chen KG, Mallon BS, McKay RDG, Robey PG. Human pluripotent stem cell culture: considerations for maintenance, expansion, and therapeutics. Cell Stem Cell 2014; 14:13-26. [PMID: 24388173 PMCID: PMC3915741 DOI: 10.1016/j.stem.2013.12.005] [Citation(s) in RCA: 247] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human pluripotent stem cells (hPSCs) provide powerful resources for application in regenerative medicine and pharmaceutical development. In the past decade, various methods have been developed for large-scale hPSC culture that rely on combined use of multiple growth components, including media containing various growth factors, extracellular matrices, 3D environmental cues, and modes of multicellular association. In this Protocol Review, we dissect these growth components by comparing cell culture methods and identifying the benefits and pitfalls associated with each one. We further provide criteria, considerations, and suggestions to achieve optimal cell growth for hPSC expansion, differentiation, and use in future therapeutic applications.
Collapse
Affiliation(s)
- Kevin G Chen
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara S Mallon
- NIH Stem Cell Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald D G McKay
- The Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
31
|
Titmarsh DM, Chen H, Glass NR, Cooper-White JJ. Concise review: microfluidic technology platforms: poised to accelerate development and translation of stem cell-derived therapies. Stem Cells Transl Med 2013; 3:81-90. [PMID: 24311699 DOI: 10.5966/sctm.2013-0118] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Stem cells are a powerful resource for producing a variety of cell types with utility in clinically associated applications, including preclinical drug screening and development, disease and developmental modeling, and regenerative medicine. Regardless of the type of stem cell, substantial barriers to clinical translation still exist and must be overcome to realize full clinical potential. These barriers span processes including cell isolation, expansion, and differentiation; purification, quality control, and therapeutic efficacy and safety; and the economic viability of bioprocesses for production of functional cell products. Microfluidic systems have been developed for a myriad of biological applications and have the intrinsic capability of controlling and interrogating the cellular microenvironment with unrivalled precision; therefore, they have particular relevance to overcoming such barriers to translation. Development of microfluidic technologies increasingly utilizes stem cells, addresses stem cell-relevant biological phenomena, and aligns capabilities with translational challenges and goals. In this concise review, we describe how microfluidic technologies can contribute to the translation of stem cell research outcomes, and we provide an update on innovative research efforts in this area. This timely convergence of stem cell translational challenges and microfluidic capabilities means that there is now an opportunity for both disciplines to benefit from increased interaction.
Collapse
Affiliation(s)
- Drew M Titmarsh
- Australian Institute for Bioengineering and Nanotechnology and
| | | | | | | |
Collapse
|
32
|
Kamei KI. Cutting-Edge Microfabricated Biomedical Tools for Human Pluripotent Stem Cell Research. ACTA ACUST UNITED AC 2013; 18:469-81. [DOI: 10.1177/2211068213495394] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
33
|
Yoshimitsu R, Hattori K, Sugiura S, Kondo Y, Yamada R, Tachikawa S, Satoh T, Kurisaki A, Ohnuma K, Asashima M, Kanamori T. Microfluidic perfusion culture of human induced pluripotent stem cells under fully defined culture conditions. Biotechnol Bioeng 2013; 111:937-47. [PMID: 24222619 DOI: 10.1002/bit.25150] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Revised: 10/09/2013] [Accepted: 11/08/2013] [Indexed: 01/18/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) are a promising cell source for drug screening. For this application, self-renewal or differentiation of the cells is required, and undefined factors in the culture conditions are not desirable. Microfluidic perfusion culture allows the production of small volume cultures with precisely controlled microenvironments, and is applicable to high-throughput cellular environment screening. Here, we developed a microfluidic perfusion culture system for hiPSCs that uses a microchamber array chip under defined extracellular matrix (ECM) and culture medium conditions. By screening various ECMs we determined that fibronectin and laminin are appropriate for microfluidic devices made out of the most popular material, polydimethylsiloxane (PDMS). We found that the growth rate of hiPSCs under pressure-driven perfusion culture conditions was higher than under static culture conditions in the microchamber array. We applied our new system to self-renewal and differentiation cultures of hiPSCs, and immunocytochemical analysis showed that the state of the hiPSCs was successfully controlled. The effects of three antitumor drugs on hiPSCs were comparable between microchamber array and 96-well plates. We believe that our system will be a platform technology for future large-scale screening of fully defined conditions for differentiation cultures on integrated microfluidic devices.
Collapse
Affiliation(s)
- Ryosuke Yoshimitsu
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata, 940-2188, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Tarn MD, Lopez-Martinez MJ, Pamme N. On-chip processing of particles and cells via multilaminar flow streams. Anal Bioanal Chem 2013; 406:139-61. [DOI: 10.1007/s00216-013-7363-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Mark D Tarn
- Department of Chemistry, The University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | | | | |
Collapse
|
36
|
Kamei KI, Hirai Y, Tabata O. Body on a Chip: Re-Creation of a Living System In Vitro. IEEE NANOTECHNOLOGY MAGAZINE 2013. [DOI: 10.1109/mnano.2013.2275024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
Lee CY, Romanova EV, Sweedler JV. Laminar stream of detergents for subcellular neurite damage in a microfluidic device: a simple tool for the study of neuroregeneration. J Neural Eng 2013; 10:036020. [PMID: 23656702 DOI: 10.1088/1741-2560/10/3/036020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The regeneration and repair of damaged neuronal networks is a difficult process to study in vivo, leading to the development of multiple in vitro models and techniques for studying nerve injury. Here we describe an approach for generating a well-defined subcellular neurite injury in a microfluidic device. APPROACH A defined laminar stream of sodium dodecyl sulfate (SDS) was used to damage selected portions of neurites of individual neurons. The somata and neurites unaffected by the SDS stream remained viable, thereby enabling the study of neuronal regeneration. MAIN RESULTS By using well-characterized neurons from Aplysia californica cultured in vitro, we demonstrate that our approach is useful in creating neurite damage, investigating neurotrophic factors, and monitoring somata migration during regeneration. Supplementing the culture medium with acetylcholinesterase (AChE) or Aplysia hemolymph facilitated the regeneration of the peptidergic Aplysia neurons within 72 h, with longer (p < 0.05) and more branched (p < 0.05) neurites than in the control medium. After the neurons were transected, their somata migrated; intriguingly, for the control cultures, the migration direction was always away from the injury site (7/7). In the supplemented cultures, the number decreased to 6/8 in AChE and 4/8 in hemolymph, with reduced migration distances in both cases. SIGNIFICANCE The SDS transection approach is simple and inexpensive, yet provides flexibility in studying neuroregeneration, particularly when it is important to make sure there are no retrograde signals from the distal segments affecting regeneration. Neurons are known to not only be under tension but also balanced in terms of force, and the balance is obviously disrupted by transection. Our experimental platform, verified with Aplysia, can be extended to mammalian systems, and help us gain insight into the role that neurotrophic factors and mechanical tension play during neuronal regeneration.
Collapse
Affiliation(s)
- Chang Young Lee
- School of Nano-Bioscience and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| | | | | |
Collapse
|
38
|
Keenan TM, Grinager JR, Procak AA, Svendsen CN. In vitro localization of human neural stem cell neurogenesis by engineered FGF-2 gradients. Integr Biol (Camb) 2013; 4:1522-31. [PMID: 23147909 DOI: 10.1039/c2ib20074k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The development of effective stem cell-based therapies for treating brain disorders is keenly dependent upon an understanding of how to generate specific neural cell types and organize them into functional, higher-order tissues analogous to those of the cerebral cortex. Studies of cortical development have revealed that the proper formation of the human cerebral cortex results from specific intercellular interactions and soluble signaling between the highly-proliferative region occupied by dividing neural stem cells and an adjacent region of active neurogenesis and neural migration. However, the factors responsible for establishing this key asymmetrical proliferative-neurogenic architecture are not entirely known. Fibroblast growth factor 2 (FGF-2) is observed in a ventricular-pial gradient during in vivo development and has been previously shown to have effects on both human neural stem cell (hNSC) proliferation and neurogenesis. Here we have adapted a microfluidic approach for creating stable concentration gradients in 3D hydrogels to explore whether FGF-2 gradients can establish defined regions of proliferation and neurogenesis in hNSC cultures. Exponential but not linear FGF-2 gradients between 0-2 ng mL(-1) were able to preferentially boost the percentage of TuJ1(+) neurons in the low concentration regions of the gradient and at levels significantly higher than in non-gradient controls. However, no gradient-dependent localization was observed for dividing hNSCs or hNSC-derived intermediate progenitors. These data suggest that exponential FGF2 gradients are useful for generating asymmetric neuron cultures, but require contributions from other factors to recapitulate the highly-proliferative ventricular zone niche. The relevance of the findings of this study to in vivo cortical development must be more cautiously stated given the artifactual nature of hNSCs and the inability of any in vitro system to fully recapitulate the chemical complexity of the developing cortex. However, it is quite possible that exponential FGF2 gradients are employed in vivo to establish or maintain an asymmetric distribution of neurons in the ventricular-pial axis of the developing cerebral cortex.
Collapse
Affiliation(s)
- T M Keenan
- Stem Cell and Regenerative Medicine Center, University of Wisconsin, 1111 Highland Ave., Madison, WI 53705, USA.
| | | | | | | |
Collapse
|
39
|
Pai JH, Kluckman K, Cowley DO, Bortner DM, Sims CE, Allbritton NL, Allbritton NL. Efficient division and sampling of cell colonies using microcup arrays. Analyst 2013; 138:220-8. [PMID: 23099535 PMCID: PMC3509232 DOI: 10.1039/c2an36065a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A microengineered array to sample clonal colonies is described. The cells were cultured on an array of individually releasable elements until the colonies expanded to cover multiple elements. Single elements were released using a laser-based system and collected to sample cells from individual colonies. A greater than an 85% rate in splitting and collecting colonies was achieved using a 3-dimensional cup-like design or "microcup". Surface modification using patterned titanium deposition of the glass substrate improved the stability of microcup adhesion to the glass while enabling minimization of the laser energy for splitting the colonies. Smaller microcup dimensions and slotting the microcup walls reduced the time needed for colonies to expand into multiple microcups. The stem cell colony retained on the array and the collected fraction within released microcups remained undifferentiated and viable. The colony samples were characterized by both reporter gene expression and a destructive assay (PCR) to identify target colonies. The platform is envisioned as a means to rapidly establish cell lines using a destructive assay to identify desired clones.
Collapse
Affiliation(s)
- Jeng-Hao Pai
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, Fax: +1 (919) 962-2388, Tel: +1 (919) 966-2291
| | | | - Dale O. Cowley
- TransViragen, Inc., PO Box 110301, Research Triangle Park, NC 27709
| | - Donna M. Bortner
- TransViragen, Inc., PO Box 110301, Research Triangle Park, NC 27709
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, Fax: +1 (919) 962-2388, Tel: +1 (919) 966-2291
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, Fax: +1 (919) 962-2388, Tel: +1 (919) 966-2291
| | - Nancy L. Allbritton
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
40
|
Tehranirokh M, Kouzani AZ, Francis PS, Kanwar JR. Microfluidic devices for cell cultivation and proliferation. BIOMICROFLUIDICS 2013; 7:51502. [PMID: 24273628 PMCID: PMC3829894 DOI: 10.1063/1.4826935] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/24/2013] [Indexed: 05/07/2023]
Abstract
Microfluidic technology provides precise, controlled-environment, cost-effective, compact, integrated, and high-throughput microsystems that are promising substitutes for conventional biological laboratory methods. In recent years, microfluidic cell culture devices have been used for applications such as tissue engineering, diagnostics, drug screening, immunology, cancer studies, stem cell proliferation and differentiation, and neurite guidance. Microfluidic technology allows dynamic cell culture in microperfusion systems to deliver continuous nutrient supplies for long term cell culture. It offers many opportunities to mimic the cell-cell and cell-extracellular matrix interactions of tissues by creating gradient concentrations of biochemical signals such as growth factors, chemokines, and hormones. Other applications of cell cultivation in microfluidic systems include high resolution cell patterning on a modified substrate with adhesive patterns and the reconstruction of complicated tissue architectures. In this review, recent advances in microfluidic platforms for cell culturing and proliferation, for both simple monolayer (2D) cell seeding processes and 3D configurations as accurate models of in vivo conditions, are examined.
Collapse
|
41
|
Reichen M, Macown RJ, Jaccard N, Super A, Ruban L, Griffin LD, Veraitch FS, Szita N. Microfabricated modular scale-down device for regenerative medicine process development. PLoS One 2012; 7:e52246. [PMID: 23284952 PMCID: PMC3526573 DOI: 10.1371/journal.pone.0052246] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 11/16/2012] [Indexed: 01/09/2023] Open
Abstract
The capacity of milli and micro litre bioreactors to accelerate process development has been successfully demonstrated in traditional biotechnology. However, for regenerative medicine present smaller scale culture methods cannot cope with the wide range of processing variables that need to be evaluated. Existing microfabricated culture devices, which could test different culture variables with a minimum amount of resources (e.g. expensive culture medium), are typically not designed with process development in mind. We present a novel, autoclavable, and microfabricated scale-down device designed for regenerative medicine process development. The microfabricated device contains a re-sealable culture chamber that facilitates use of standard culture protocols, creating a link with traditional small-scale culture devices for validation and scale-up studies. Further, the modular design can easily accommodate investigation of different culture substrate/extra-cellular matrix combinations. Inactivated mouse embryonic fibroblasts (iMEF) and human embryonic stem cell (hESC) colonies were successfully seeded on gelatine-coated tissue culture polystyrene (TC-PS) using standard static seeding protocols. The microfluidic chip included in the device offers precise and accurate control over the culture medium flow rate and resulting shear stresses in the device. Cells were cultured for two days with media perfused at 300 µl.h−1 resulting in a modelled shear stress of 1.1×10−4 Pa. Following perfusion, hESC colonies stained positively for different pluripotency markers and retained an undifferentiated morphology. An image processing algorithm was developed which permits quantification of co-cultured colony-forming cells from phase contrast microscope images. hESC colony sizes were quantified against the background of the feeder cells (iMEF) in less than 45 seconds for high-resolution images, which will permit real-time monitoring of culture progress in future experiments. The presented device is a first step to harness the advantages of microfluidics for regenerative medicine process development.
Collapse
Affiliation(s)
- Marcel Reichen
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Rhys J. Macown
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Nicolas Jaccard
- Department of Biochemical Engineering, University College London, London, United Kingdom
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, United Kingdom
| | - Alexandre Super
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Ludmila Ruban
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Lewis D. Griffin
- Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, United Kingdom
- Department of Computer Science, University College London, London, United Kingdom
| | - Farlan S. Veraitch
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
42
|
Gach PC, Xu W, King SJ, Sims CE, Bear J, Allbritton NL. Microfabricated arrays for splitting and assay of clonal colonies. Anal Chem 2012; 84:10614-20. [PMID: 23153031 PMCID: PMC3525785 DOI: 10.1021/ac301895t] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A microfabricated platform was developed for highly parallel and efficient colony picking, splitting, and clone identification. A pallet array provided patterned cell colonies which mated to a second printing array composed of bridging microstructures formed by a supporting base and attached post. The posts enabled mammalian cells from colonies initially cultured on the pallet array to migrate to corresponding sites on the printing array. Separation of the arrays simultaneously split the colonies, creating a patterned replica. Optimization of array elements provided transfer efficiencies greater than 90% using bridging posts of 30 μm diameter and 100 μm length and total colony numbers of 3000. Studies using five mammalian cell lines demonstrated that a variety of adherent cell types could be cultured and effectively split with printing efficiencies of 78-92%. To demonstrate the technique's utility, clonal cell lines with siRNA knockdown of Coronin 1B were generated using the arrays and compared to a traditional FACS/Western Blotting-based approach. Identification of target clones required a destructive assay to identify cells with an absence of Coronin 1B brought about by the successful infection of interfering shRNA construct. By virtue of miniaturization and its parallel format, the platform enabled the identification and generation of 12 target clones from a starting sample of only 3900 cells and required only 5 man hours over 11 days. In contrast, the traditional method required 500,000 cells and generated only 5 target clones with 34 man hours expended over 47 days. These data support the considerable reduction in time, manpower, and reagents using the miniaturized platform for clonal selection by destructive assay versus conventional approaches.
Collapse
Affiliation(s)
- Philip C. Gach
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Wei Xu
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Samantha J. King
- Department of Cell & Development Biology, University of North Carolina, Chapel Hill, NC 27599
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - James Bear
- Department of Cell & Development Biology, University of North Carolina, Chapel Hill, NC 27599
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599 and North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
43
|
Kawada J, Kimura H, Akutsu H, Sakai Y, Fujii T. Spatiotemporally controlled delivery of soluble factors for stem cell differentiation. LAB ON A CHIP 2012; 12:4508-15. [PMID: 22968416 DOI: 10.1039/c2lc40268h] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Despite the fact that cells in vivo are largely affected by the spatial heterogeneity in their surroundings, in vitro experimental procedures for stem cell differentiation have been relying on spatially uniform culture environments so far. Here, we present a method to form spatiotemporally non-uniform culture environments for stem cell differentiation using a membrane-based microfluidic device. By adopting a porous membrane with relatively large pores, patterned delivery of soluble factors is maintained stably over a period of time long enough for cell differentiation. We report that spatial patterns of mouse induced pluripotent stem cells (miPSCs) differentiation can be controlled by the present method. Furthermore, it is shown that the cell fate decision of miPSCs is determined by time-dependent switching of the delivery pattern. The present technique could be of relevance to the detailed analyses of the characteristics of stem cell differentiation in time and space, opening up a new insight into regenerative biology.
Collapse
Affiliation(s)
- Jiro Kawada
- Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
44
|
Mortato M, Blasi L, Barbarella G, Argentiere S, Gigli G. pH controlled staining of CD4(+) and CD19(+) cells within functionalized microfluidic channel. BIOMICROFLUIDICS 2012; 6:44107. [PMID: 24191176 PMCID: PMC3505194 DOI: 10.1063/1.4763560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 10/10/2012] [Indexed: 05/31/2023]
Abstract
Herein proposed is a simple system to realize hands-free labeling and simultaneous detection of two human cell lines within a microfluidic device. This system was realized by novel covalent immobilization of pH-responsive poly(methacrylic acid) microgels onto the inner glass surface of an assembled polydimethylsiloxane/glass microfluidic channel. Afterwards, selected thiophene labeled monoclonal antibodies, specific for recognition of CD4 antigens on T helper/inducer cells and CD19 antigens on B lymphocytes cell lines, were encapsulated in their active state by the immobilized microgels. When the lymphocytes suspension, containing the two target subpopulations, was flowed through the microchannel, the physiological pH of the cellular suspension induced the release of the labeled antibodies from the microgels and thus the selective cellular staining. The selective pH-triggered staining of the CD4- and CD19-positive cells was investigated in this preliminary experimental study by laser scanning confocal microscopy. This approach represents an interesting and versatile tool to realize cellular staining in a defined module of lab-on-a-chip devices for subsequent detection and counting.
Collapse
Affiliation(s)
- Mariangela Mortato
- Superior School ISUFI, University of Salento, via Arnesano, I-73100 Lecce, Italy ; NNL CNR-Institute of Nanoscience, via Arnesano, I-73100 Lecce, Italy
| | | | | | | | | |
Collapse
|
45
|
Lesher-Perez SC, Frampton JP, Takayama S. Microfluidic systems: a new toolbox for pluripotent stem cells. Biotechnol J 2012; 8:180-91. [PMID: 23125055 DOI: 10.1002/biot.201200206] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/23/2012] [Accepted: 09/25/2012] [Indexed: 01/09/2023]
Abstract
Conventional culture systems are often limited in their ability to regulate the growth and differentiation of pluripotent stem cells. Microfluidic systems can overcome some of these limitations by providing defined growth conditions with user-controlled spatiotemporal cues. Microfluidic systems allow researchers to modulate pluripotent stem cell renewal and differentiation through biochemical and mechanical stimulation, as well as through microscale patterning and organization of cells and extracellular materials. Essentially, microfluidic tools are reducing the gap between in vitro cell culture environments and the complex and dynamic features of the in vivo stem cell niche. These microfluidic culture systems can also be integrated with microanalytical tools to assess the health and molecular status of pluripotent stem cells. The ability to control biochemical and mechanical input to cells, as well as rapidly and efficiently analyze the biological output from cells, will further our understanding of stem cells and help translate them into clinical use. This review provides a comprehensive insignt into the implications of microfluidics on pluripotent stem cell research.
Collapse
|
46
|
Liu Y, Butler WB, Pappas D. Spatially selective reagent delivery into cancer cells using a two-layer microfluidic culture system. Anal Chim Acta 2012; 743:125-30. [PMID: 22882832 PMCID: PMC3439837 DOI: 10.1016/j.aca.2012.06.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 06/25/2012] [Accepted: 06/29/2012] [Indexed: 12/31/2022]
Abstract
In this work, we demonstrate a two-layer microfluidic system capable of spatially selective delivery of drugs and other reagents under low shear stress. Loading occurs by hydrodynamically focusing a reagent stream over a particular region of the cell culture. The system consisted of a cell culture chamber and fluid flow channel, which were located in different layers to reduce shear stress on cells. Cells in the center of the culture chamber were exposed to parallel streams of laminar flow, which allowed fast changes to be made to the cellular environment. The shear force was reduced to 2.7 dyn cm(-2) in the two-layer device (vs. 6.0 dyn cm(-2) in a one-layer device). Cells in the side of the culture chamber were exposed to the side streams of buffer; the shear force was further reduced to a greater extent since the sides of the culture chamber were separated from the main fluid path. The channel shape and flow rate of the multiple streams were optimized for spatially controlled reagent delivery. The boundaries between streams were well controlled at a flow rate of 0.1 mL h(-1), which was optimized for all streams. We demonstrated multi-reagent delivery to different regions of the same culture well, as well as selective treatment of cancer cells with a built in control group in the same well. In the case of apoptosis induction using staurosporine, 10% of cells remained viable after 24 h of exposure. Cells in the same chamber, but not exposed to staurosporine, had a viability of 90%. This chip allows dynamic observation of cellular behavior immediately after drug delivery, as well as long-term drug treatment with the benefit of large cell numbers, device simplicity, and low shear stress.
Collapse
Affiliation(s)
- Yan Liu
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| | - W. Boyd Butler
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409
| | - Dimitri Pappas
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409
| |
Collapse
|
47
|
Gao D, Liu H, Jiang Y, Lin JM, Gao D, Liu H, Jiang Y. Recent developments in microfluidic devices for in vitro cell culture for cell-biology research. Trends Analyt Chem 2012. [DOI: 10.1016/j.trac.2012.02.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
48
|
Frampton JP, Lai D, Sriram H, Takayama S. Precisely targeted delivery of cells and biomolecules within microchannels using aqueous two-phase systems. Biomed Microdevices 2012; 13:1043-51. [PMID: 21769637 DOI: 10.1007/s10544-011-9574-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Laminar and pulsatile flow of aqueous solutions in microfluidic channels can be useful for controlled delivery of cells and molecules. Dispersion effects resulting from diffusion and convective disturbances, however, result in reagent delivery profiles becoming blurred over the length of the channels. This issue is addressed partially by using oil-in-water phase systems. However, there are limitations in terms of the biocompatibility of these systems for adherent cell culture. Here we present a fully biocompatible aqueous two-phase flow system that can be used to pattern cells within simple microfluidic channel designs, as well as to deliver biochemical treatments to cells according to discrete boundaries. We demonstrate that aqueous two-phase systems are capable of precisely delivering cells as laminar patterns, or as islands by way of forced droplet formation. We also demonstrate that these systems can be used to precisely control chemical delivery to preformed monolayers of cells growing within channels. Treatments containing trypsin were localized more reliably using aqueous two-phase delivery than using conventional delivery in aqueous medium.
Collapse
Affiliation(s)
- John P Frampton
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | |
Collapse
|
49
|
Smith GD, Takayama S, Swain JE. Rethinking in vitro embryo culture: new developments in culture platforms and potential to improve assisted reproductive technologies. Biol Reprod 2012; 86:62. [PMID: 21998170 DOI: 10.1095/biolreprod.111.095778] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The preponderance of research toward improving embryo development in vitro has focused on manipulation of the chemical soluble environment, including altering basic salt composition, energy substrate concentration, amino acid makeup, and the effect of various growth factors or addition or subtraction of other supplements. In contrast, relatively little work has been done examining the physical requirements of preimplantation embryos and the role culture platforms or devices can play in influencing embryo development within the laboratory. The goal of this review is not to reevaluate the soluble composition of past and current embryo culture media, but rather to consider how other controlled and precise factors such as time, space, mechanical interactions, gradient diffusions, cell movement, and surface interactions might influence embryo development. Novel culture platforms are being developed as a result of interdisciplinary collaborations between biologists and biomedical, material, chemical, and mechanical engineers. These approaches are looking beyond the soluble media composition and examining issues such as media volume and embryo spacing. Furthermore, methods that permit precise and regulated dynamic embryo culture with fluid flow and embryo movement are now available, and novel culture surfaces are being developed and tested. While several factors remain to be investigated to optimize the efficiency of embryo production, manipulation of the embryo culture microenvironment through novel devices and platforms may offer a pathway toward improving embryo development within the laboratory of the future.
Collapse
Affiliation(s)
- Gary D Smith
- Department of Obstetrics & Gynecology, University of Michigan, Ann Arbor, Michigan 48109-0617, USA.
| | | | | |
Collapse
|
50
|
Przybyla L, Voldman J. Probing embryonic stem cell autocrine and paracrine signaling using microfluidics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2012; 5:293-315. [PMID: 22524217 PMCID: PMC4030416 DOI: 10.1146/annurev-anchem-062011-143122] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Although stem cell fate is traditionally manipulated by exogenously altering the cells' extracellular signaling environment, the endogenous autocrine and paracrine signals produced by the cells also contribute to their two essential processes: self-renewal and differentiation. Autocrine and/or paracrine signals are fundamental to both embryonic stem cell self-renewal and early embryonic development, but the nature and contributions of these signals are often difficult to fully define using conventional methods. Microfluidic techniques have been used to explore the effects of cell-secreted signals by controlling cell organization or by providing precise control over the spatial and temporal cellular microenvironment. Here we review how such techniques have begun to be adapted for use with embryonic stem cells, and we illustrate how many remaining questions in embryonic stem cell biology could be addressed using microfluidic technologies.
Collapse
Affiliation(s)
- Laralynne Przybyla
- Dept. of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA, 02139
| | - Joel Voldman
- Dept. Of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, USA, 02139
| |
Collapse
|