1
|
Kim K, Lee Y, Jung KB, Kim Y, Jang E, Lee MO, Son MY, Lee HJ. Highly Stretchable 3D Microelectrode Array for Noninvasive Functional Evaluation of Cardiac Spheroids and Midbrain Organoids. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2412953. [PMID: 39676473 DOI: 10.1002/adma.202412953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/08/2024] [Indexed: 12/17/2024]
Abstract
Organoids are 3D biological models that recapitulate the complex structures and functions of human organs. Despite the rapid growth in the generation of organoids, in vitro assay tools are still limited to 2D forms. Thus, a comprehensive and continuous functional evaluation of the electrogenic organoids remains a challenge. Here, a highly stretchable 3D multielectrode array (sMEA) with protruding microelectrodes is presented for functional evaluation of electrogenic organoids. The optimized serpentine structures with bridge structures cover the surface of the organoids conformally even in immersion. The protruding microelectrodes form a stable contact with the organoids and allow electrophysiological recordings with high signal-to-noise ratio (SNR). sMEAs are fabricated in wafer-scale for repeatable, scalable, and mass production and packed into an easy-to-use, user-friendly, and robust microwell for fast dissemination of technology. The versatility of sMEA is validated by measuring electrophysiological signals from cardiac spheroids and midbrain organoids with a wide range of sizes from 500 to 1500 µm. Also, electrophysiological signals recorded with high SNR enable functional evaluation of the effects of drugs. The proposed sMEA with high SNR and user-friendly interface could be the key player in high-throughput drug screening, 3D spatiotemporal mapping of electrogenic organoids, and standardization of protocols for quality assessment.
Collapse
Affiliation(s)
- Kiup Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Youngsun Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Kwang Bo Jung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Yoojeong Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Eunyoung Jang
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Mi-Ok Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Mi-Young Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyunjoo J Lee
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- KAIST Institute for NanoCentury (KINC), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
2
|
Yang Q, Xiao Z, Lv X, Zhang T, Liu H. Fabrication and Biomedical Applications of Heart-on-a-chip. Int J Bioprint 2021; 7:370. [PMID: 34286153 PMCID: PMC8287510 DOI: 10.18063/ijb.v7i3.370] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/25/2021] [Indexed: 12/26/2022] Open
Abstract
Heart diseases have become the main killer threatening human health, and various methods have been developed to study heart disease. Among them, heart-on-a-chip has emerged in recent years as a method for constructing disease (or normal) models in vitro and is considered as a promising tool to study heart diseases. Compared with other methods, the advantages of heart-on-a-chip include the high portability, high throughput, and the capability to mimic microenvironments in vivo. It has shown a great potential in disease mechanism study and drug screening. In this paper, we review the recent advances in heart-on-a-chip, including the fabrication methods (e.g., 3D bioprinting) and biomedical applications. By analyzing the structure of the existing heart-on-a-chip, we proposed that a highly integrated heart-on-a-chip includes four elements: Microfluidic chips, cells/microtissues, microactuators to construct the microenvironment, and microsensors for results readout. Finally, the current challenges and future directions of heart-on-a-chip are discussed.
Collapse
Affiliation(s)
- Qingzhen Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Micro-/Nano-technology Research Center, State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Research Institute of Xi’an Jiaotong University, Hangzhou, Zhejiang 311215, P.R. China
| | - Zhanfeng Xiao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
| | - Xuemeng Lv
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, Shaanxi 710049, P.R. China
| | - Tingting Zhang
- College of Mechanical and Electrical Engineering, Shaanxi University of Science and Technology, Xi’an, Shaanxi 710021, P.R. China
| | - Han Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province and Education Ministry of P.R. China, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450016, P.R. China
| |
Collapse
|
3
|
Pointon A, Maher J, Davis M, Baker T, Cichocki J, Ramsden D, Hale C, Kolaja KL, Levesque P, Sura R, Stresser DM, Gintant G. Cardiovascular microphysiological systems (CVMPS) for safety studies - a pharma perspective. LAB ON A CHIP 2021; 21:458-472. [PMID: 33471007 DOI: 10.1039/d0lc01040e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The integrative responses of the cardiovascular (CV) system are essential for maintaining blood flow to provide oxygenation, nutrients, and waste removal for the entire body. Progress has been made in independently developing simple in vitro models of two primary components of the CV system, namely the heart (using induced pluripotent stem-cell derived cardiomyocytes) and the vasculature (using endothelial cells and smooth muscle cells). These two in vitro biomimics are often described as immature and simplistic, and typically lack the structural complexity of native tissues. Despite these limitations, they have proven useful for specific "fit for purpose" applications, including early safety screening. More complex in vitro models offer the tantalizing prospect of greater refinement in risk assessments. To this end, efforts to physically link cardiac and vascular components to mimic a true CV microphysiological system (CVMPS) are ongoing, with the goal of providing a more holistic and integrated CV response model. The challenges of building and implementing CVMPS in future pharmacological safety studies are many, and include a) the need for more complex (and hence mature) cell types and tissues, b) the need for more realistic vasculature (within and across co-modeled tissues), and c) the need to meaningfully couple these two components to allow for integrated CV responses. Initial success will likely come with simple, bioengineered tissue models coupled with fluidics intended to mirror a vascular component. While the development of more complex integrated CVMPS models that are capable of differentiating safe compounds and providing mechanistic evaluations of CV liabilities may be feasible, adoption by pharma will ultimately hinge on model efficiency, experimental reproducibility, and added value above current strategies.
Collapse
Affiliation(s)
- Amy Pointon
- Functional Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jonathan Maher
- Translational Safety Sciences, Theravance Biopharma, South San Francisco, CA 94080, USA
| | - Myrtle Davis
- Discovery Toxicology, Bristol-Myers Squibb Company, 3553 Lawrenceville Rd Princeton, NJ 08540, USA
| | - Thomas Baker
- Eli Lilly, Lilly Corporate Center, Indianapolis IN 46285, USA
| | | | - Diane Ramsden
- Takeda Pharmaceuticals, 35 Landsdowne St., Cambridge, MA 02139, UK
| | - Christopher Hale
- Amgen Research, 1120 Veterans Blvd., S. San Francisco, 94080, USA
| | - Kyle L Kolaja
- Investigative Toxicology and Cell Therapy, Bristol-Myers Squibb Company, 556 Morris Avenue, Summit NJ 07042, USA
| | - Paul Levesque
- Discovery Toxicology, Bristol-Myers Squibb Company, 3553 Lawrenceville Rd Princeton, NJ 08540, USA
| | | | - David M Stresser
- Drug Metabolism, Pharmacokinetics and Translational Modeling, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA
| | - Gary Gintant
- Integrative Pharmacology, Integrated Science and Technology, AbbVie, 1 Waukegan Rd, N Chicago, IL 60064, USA.
| |
Collapse
|
4
|
Zuppinger C. 3D Cardiac Cell Culture: A Critical Review of Current Technologies and Applications. Front Cardiovasc Med 2019; 6:87. [PMID: 31294032 PMCID: PMC6606697 DOI: 10.3389/fcvm.2019.00087] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/10/2019] [Indexed: 12/23/2022] Open
Abstract
Three-dimensional (3D) cell culture is often mentioned in the context of regenerative medicine, for example, for the replacement of ischemic myocardium with tissue-engineered muscle constructs. Additionally, 3D cell culture is used, although less commonly, in basic research, toxicology, and drug development. These applications have recently benefited from innovations in stem cell technologies allowing the mass-production of hiPSC-derived cardiomyocytes or other cardiovascular cells, and from new culturing methods including organ-on-chip and bioprinting technologies. On the analysis side, improved sensors, computer-assisted image analysis, and data collection techniques have lowered the bar for switching to 3D cell culture models. Nevertheless, 3D cell culture is not as widespread or standardized as traditional cell culture methods using monolayers of cells on flat surfaces. The many possibilities of 3D cell culture, but also its limitations, drawbacks and methodological pitfalls, are less well-known. This article reviews currently used cardiovascular 3D cell culture production methods and analysis techniques for the investigation of cardiotoxicity, in drug development and for disease modeling.
Collapse
Affiliation(s)
- Christian Zuppinger
- Cardiology, Department of Biomedical Research, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
5
|
Bai Y, Yeung E, Lui C, Ong CS, Pitaktong I, Huang C, Inoue T, Matsushita H, Ma C, Hibino N. A Net Mold-based Method of Scaffold-free Three-Dimensional Cardiac Tissue Creation. J Vis Exp 2018. [PMID: 30124650 DOI: 10.3791/58252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
This protocol describes a novel and easy net mold-based method to create three-dimensional (3-D) cardiac tissues without additional scaffold material. Human-induced pluripotent stem-cell-derived cardiomyocytes (iPSC-CMs), human cardiac fibroblasts (HCFs), and human umbilical vein endothelial cells (HUVECs) are isolated and used to generate a cell suspension with 70% iPSC-CMs, 15% HCFs, and 15% HUVECs. They are co-cultured in an ultra-low attachment "hanging drop" system, which contains micropores for condensing hundreds of spheroids at one time. The cells aggregate and spontaneously form beating spheroids after 3 days of co-culture. The spheroids are harvested, seeded into a novel mold cavity, and cultured on a shaker in the incubator. The spheroids become a mature functional tissue approximately 7 days after seeding. The resultant multilayered tissues consist of fused spheroids with satisfactory structural integrity and synchronous beating behavior. This new method has promising potential as a reproducible and cost-effective method to create engineered tissues for the treatment of heart failure in the future.
Collapse
Affiliation(s)
- Yang Bai
- Department of Cardiac Surgery, The First Hospital of Jilin University; Division of Cardiac Surgery, Johns Hopkins Hospital
| | - Enoch Yeung
- Division of Cardiac Surgery, Johns Hopkins Hospital
| | - Cecillia Lui
- Division of Cardiac Surgery, Johns Hopkins Hospital
| | | | | | - Chenyu Huang
- Division of Cardiac Surgery, Johns Hopkins Hospital
| | | | | | - Chunye Ma
- Division of Cardiac Surgery, Johns Hopkins Hospital
| | | |
Collapse
|
6
|
Schmitz A, Fischer SC, Mattheyer C, Pampaloni F, Stelzer EHK. Multiscale image analysis reveals structural heterogeneity of the cell microenvironment in homotypic spheroids. Sci Rep 2017; 7:43693. [PMID: 28255161 PMCID: PMC5334646 DOI: 10.1038/srep43693] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/30/2017] [Indexed: 12/31/2022] Open
Abstract
Three-dimensional multicellular aggregates such as spheroids provide reliable in vitro substitutes for tissues. Quantitative characterization of spheroids at the cellular level is fundamental. We present the first pipeline that provides three-dimensional, high-quality images of intact spheroids at cellular resolution and a comprehensive image analysis that completes traditional image segmentation by algorithms from other fields. The pipeline combines light sheet-based fluorescence microscopy of optically cleared spheroids with automated nuclei segmentation (F score: 0.88) and concepts from graph analysis and computational topology. Incorporating cell graphs and alpha shapes provided more than 30 features of individual nuclei, the cellular neighborhood and the spheroid morphology. The application of our pipeline to a set of breast carcinoma spheroids revealed two concentric layers of different cell density for more than 30,000 cells. The thickness of the outer cell layer depends on a spheroid’s size and varies between 50% and 75% of its radius. In differently-sized spheroids, we detected patches of different cell densities ranging from 5 × 105 to 1 × 106 cells/mm3. Since cell density affects cell behavior in tissues, structural heterogeneities need to be incorporated into existing models. Our image analysis pipeline provides a multiscale approach to obtain the relevant data for a system-level understanding of tissue architecture.
Collapse
Affiliation(s)
- Alexander Schmitz
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15 - D-60348 Frankfurt am Main, Germany
| | - Sabine C Fischer
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15 - D-60348 Frankfurt am Main, Germany
| | - Christian Mattheyer
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15 - D-60348 Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15 - D-60348 Frankfurt am Main, Germany
| | - Ernst H K Stelzer
- Physical Biology/Physikalische Biologie (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt - Macromolecular Complexes (CEF - MC), Goethe Universität - Frankfurt am Main (Campus Riedberg), Max-von-Laue-Straße 15 - D-60348 Frankfurt am Main, Germany
| |
Collapse
|
7
|
Zhang YS, Arneri A, Bersini S, Shin SR, Zhu K, Goli-Malekabadi Z, Aleman J, Colosi C, Busignani F, Dell'Erba V, Bishop C, Shupe T, Demarchi D, Moretti M, Rasponi M, Dokmeci MR, Atala A, Khademhosseini A. Bioprinting 3D microfibrous scaffolds for engineering endothelialized myocardium and heart-on-a-chip. Biomaterials 2016; 110:45-59. [PMID: 27710832 DOI: 10.1016/j.biomaterials.2016.09.003] [Citation(s) in RCA: 560] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 08/30/2016] [Accepted: 09/03/2016] [Indexed: 02/06/2023]
Abstract
Engineering cardiac tissues and organ models remains a great challenge due to the hierarchical structure of the native myocardium. The need of integrating blood vessels brings additional complexity, limiting the available approaches that are suitable to produce integrated cardiovascular organoids. In this work we propose a novel hybrid strategy based on 3D bioprinting, to fabricate endothelialized myocardium. Enabled by the use of our composite bioink, endothelial cells directly bioprinted within microfibrous hydrogel scaffolds gradually migrated towards the peripheries of the microfibers to form a layer of confluent endothelium. Together with controlled anisotropy, this 3D endothelial bed was then seeded with cardiomyocytes to generate aligned myocardium capable of spontaneous and synchronous contraction. We further embedded the organoids into a specially designed microfluidic perfusion bioreactor to complete the endothelialized-myocardium-on-a-chip platform for cardiovascular toxicity evaluation. Finally, we demonstrated that such a technique could be translated to human cardiomyocytes derived from induced pluripotent stem cells to construct endothelialized human myocardium. We believe that our method for generation of endothelialized organoids fabricated through an innovative 3D bioprinting technology may find widespread applications in regenerative medicine, drug screening, and potentially disease modeling.
Collapse
Affiliation(s)
- Yu Shrike Zhang
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA.
| | - Andrea Arneri
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Simone Bersini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milan 20161, Italy
| | - Su-Ryon Shin
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA
| | - Kai Zhu
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zahra Goli-Malekabadi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biomedical Engineering, Amirkabir University of Technology, Tehran 64540, Iran
| | - Julio Aleman
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cristina Colosi
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Sapienza Università di Roma, Rome 00185, Italy
| | - Fabio Busignani
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Electronics and Telecommunications, Politecnico di Torino, Torino 10129, Italy
| | - Valeria Dell'Erba
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biomedical Engineering, Politecnico di Torino, Torino 10129, Italy
| | - Colin Bishop
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| | - Thomas Shupe
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, Torino 10129, Italy
| | - Matteo Moretti
- Cell and Tissue Engineering Lab, IRCCS Istituto Ortopedico Galeazzi, Milan 20161, Italy; Swiss Institute for Regnerative Medicine, Lugano 6900, Switzerland; Cardiocentro Ticino, Lugano 6900, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Mehmet Remzi Dokmeci
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02139, USA; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Seoul 143-701, Republic of Korea; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|
8
|
Tsui JH, Lee W, Pun SH, Kim J, Kim DH. Microfluidics-assisted in vitro drug screening and carrier production. Adv Drug Deliv Rev 2013; 65:1575-88. [PMID: 23856409 DOI: 10.1016/j.addr.2013.07.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/06/2013] [Accepted: 07/05/2013] [Indexed: 12/11/2022]
Abstract
Microfluidic platforms provide several unique advantages for drug development. In the production of drug carriers, physical properties such as size and shape, and chemical properties such as drug composition and pharmacokinetic parameters, can be modified simply and effectively by tuning the flow rate and geometries. Large numbers of carriers can then be fabricated with minimal effort and with little to no batch-to-batch variation. Additionally, cell or tissue culture models in microfluidic systems can be used as in vitro drug screening tools. Compared to in vivo animal models, microfluidic drug screening platforms allow for high-throughput and reproducible screening at a significantly lower cost, and when combined with current advances in tissue engineering, are also capable of mimicking native tissues. In this review, various microfluidic platforms for drug and gene carrier fabrication are reviewed to provide guidelines for designing appropriate carriers. In vitro microfluidic drug screening platforms designed for high-throughput analysis and replication of in vivo conditions are also reviewed to highlight future directions for drug research and development.
Collapse
Affiliation(s)
- Jonathan H Tsui
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
9
|
Gáti I, Danielsson O, Betmark T, Ernerudh J, Öllinger K, Dizdar N. Culturing of diagnostic muscle biopsies as spheroid-like structures: a pilot study of morphology and viability. Neurol Res 2013; 32:650-5. [DOI: 10.1179/016164109x12464612122579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
10
|
A scaffold-free in vitro model for osteogenesis of human mesenchymal stem cells. Tissue Cell 2011; 43:91-100. [PMID: 21329953 DOI: 10.1016/j.tice.2010.12.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 12/21/2010] [Accepted: 12/27/2010] [Indexed: 11/21/2022]
Abstract
For studying cellular processes three-dimensional (3D) in vitro models are of a high importance. For tissue engineering approaches osseous differentiation is performed on 3D scaffolds, but material depending influences promote cellular processes like adhesion, proliferation and differentiation. To investigate developmental processes of mesenchymal stem cells without cell-substrate interactions, self-contained in vitro models mimicking physiological condition are required. However, with respect to scientific investigations and pharmaceutical tests, it is essential that these tissue models are well characterised and are of a high reproducibility. In order to establish an appropriate in vitro model for bone formation, different protocols are compared and optimised regarding their aggregate formation efficiency, homogeneity of the aggregates, the viability and their ability to induce differentiation into the osteogenic lineage. The protocols for the generation of 3D cell models are based on rotation culture, hanging drop technique, and the cultivation in non adhesive culture vessels (single vessels as well as 96 well plates). To conclude, the cultivation of hMSCs in 96 well non adhesive plates facilitates an easy way to cultivate homogenous cellular aggregates with high performance efficiency in parallel. The size can be controlled by the initial cell density per well and within this spheroids, bone formation has been induced.
Collapse
|
11
|
Daus AW, Goldhammer M, Layer PG, Thielemann C. Electromagnetic exposure of scaffold-free three-dimensional cell culture systems. Bioelectromagnetics 2011; 32:351-9. [PMID: 21280061 DOI: 10.1002/bem.20649] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 12/24/2010] [Indexed: 11/11/2022]
Abstract
In recent years, a number of in vitro studies have reported on the possible athermal effects of electromagnetic exposure on biological tissue. Typically, this kind of study is performed on monolayers of primary cells or cell lines. However, two-dimensional cell layer systems lack physiological relevance since cells in vivo are organized in a three-dimensional (3D) architecture. In monolayer studies, cell-cell and cell-ECM interactions obviously differ from live tissue and scale-ups of experimental results to in vivo systems should be considered carefully. To overcome this problem, we used a scaffold-free 3D cell culture system, suitable for the exploration of electrophysiological effects due to electromagnetic fields (EMF) at 900 MHz. Dissociated cardiac myocytes were reaggregated into cellular spheres by constant rotation, and non-invasive extracellular recordings of these so-called spheroids were performed with microelectrode arrays (MEA). In this study, 3D cell culture systems were exposed to pulsed EMFs in a stripline setup. We found that inhomogeneities in the EMF due to electrodes and conducting lines of the MEA chip had only a minor influence on the field distribution in the spheroid if the exposure parameters were chosen carefully.
Collapse
Affiliation(s)
- Andreas W Daus
- Bioelectronics and BioMEMS Laboratory, University of Applied Sciences Aschaffenburg, Aschaffenburg, Germany.
| | | | | | | |
Collapse
|
12
|
Pirlo RK, Ma Z, Sweeney A, Liu H, Yun JX, Peng X, Yuan X, Guo GX, Gao BZ. Laser-guided cell micropatterning system. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2011; 82:013708. [PMID: 21280838 PMCID: PMC3045411 DOI: 10.1063/1.3529919] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Employing optical force, our laser-guided cell micropatterning system, is capable of patterning different cell types onto and within standard cell research devices, including commercially available multielectrode arrays (MEAs) with glass culture rings, 35 mm Petri dishes, and microdevices microfabricated with polydimethylsiloxane on 22 mm × 22 mm cover glasses. We discuss the theory of optical forces for generating laser guidance and the calculation of optimal beam characteristics for cell guidance. We describe the hardware design and software program for the cell patterning system. Finally, we demonstrate the capabilities of the system by (1) patterning neurons to form an arbitrary pattern, (2) patterning neurons onto the electrodes of a standard MEA, and (3) patterning and aligning adult cardiomyocytes in a polystyrene Petri dish.
Collapse
Affiliation(s)
- Russell K Pirlo
- Department of Bioengineering, COMSET, Clemson University, South Carolina 29634, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Robins JC, Morgan JR, Krueger P, Carson SA. Bioengineering anembryonic human trophoblast vesicles. Reprod Sci 2010; 18:128-35. [PMID: 20978180 DOI: 10.1177/1933719110381923] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Trophoblast cells in vivo form a 3-dimensional structure that promotes complex cell-to-cell interactions that cannot be studied with traditional monolayer culture. We describe a 3-dimensional trophoblast bioreactor to study cellular interactions. METHODS Nonadhesive agarose hydrogels were cast from molds using computer-assisted prototyping. Trophoblast cells were seeded into the gels for 10 days. Morphology, viability, and vesicle behavior were assessed. RESULTS Trophoblast cells formed uniform spheroids. Serial sectioning on days 3, 7, and 10 revealed central vacuolization with a consistent outer rim 12.3-μ thick. The vesicle configuration has been confirmed with confocal imaging. Electron Microscopic (EM) imaging revealed its ultrastructure. The vesicles migrate across a fibronectin-coated surface and invaded basement membrane. CONCLUSIONS Trophoblast cells cultured in a novel substrate-free 3-dimensional system form trophoblast vesicles. This new cell culture technique allows us to better study placental cell-to-cell interactions with the potential of forming microtissues.
Collapse
Affiliation(s)
- Jared C Robins
- Departments of Obstetrics and Gynecology, Warren Alpert Medical School at Brown University, Providence, RI 02067, USA.
| | | | | | | |
Collapse
|
14
|
Wang Y, Wong LB, Mao H. Induction of ciliated cells from avian embryonic stem cells using three-dimensional matrix. Tissue Eng Part C Methods 2010; 16:929-36. [PMID: 19951007 PMCID: PMC2963634 DOI: 10.1089/ten.tec.2009.0327] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 11/30/2009] [Indexed: 11/12/2022] Open
Abstract
We have devised a simple three-dimensional (3D) tissue-culturing method to induce ciliogenesis from avian embryonic stem (ES) cells by using avian fertilized eggs. Unlike the previous reported techniques, this method does not require trypsinization, which would reduce the viability of the cells; it also does not require an air-liquid interface to induce ciliogenesis and to maintain the growth of the induced ciliated cells. ES cells seeded and attached on this collagen-coated chitosan 3D gel grew spontaneously and robustly. Following 2 weeks in culture with inhibition of embryoid body formation, cells with noticeable and vigorous beating cilia were observed. We measured the ciliary beat frequencies of these ES-differentiated ciliated cells for 40 days. These results were consistent with all reported measurements made for other species of ciliated cells, including human, from our previous study. These data imply that the cilia of these ES-derived ciliated cells, beating at their intrinsic basal autorhythmic rate, preserve the integrity of the regulatory mechanisms of ciliary beat frequency. In conclusion, we have shown that ES cells cultured in a 3D tissue-engineered scaffold is a promising approach for developing an in vitro cell model that closely mimics the in vivo ciliated cell natural milieu. This cell model can potentially be the source of ciliated cells for cell-based high-throughput screening and discovery of pulmonary drugs.
Collapse
Affiliation(s)
- Yuchi Wang
- BioTechPlex Corporation, San Marcos, California 92078, USA.
| | | | | |
Collapse
|
15
|
Nirmalanandhan VS, Duren A, Hendricks P, Vielhauer G, Sittampalam GS. Activity of Anticancer Agents in a Three-Dimensional Cell Culture Model. Assay Drug Dev Technol 2010; 8:581-90. [DOI: 10.1089/adt.2010.0276] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Victor Sanjit Nirmalanandhan
- The Institute for Advancing Medical Innovation, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Alicia Duren
- The Institute for Advancing Medical Innovation, The University of Kansas Medical Center, Kansas City, Kansas
| | - Peter Hendricks
- The Institute for Advancing Medical Innovation, The University of Kansas Medical Center, Kansas City, Kansas
| | - George Vielhauer
- The Institute for Advancing Medical Innovation, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Urology, The University of Kansas Medical Center, Kansas City, Kansas
| | - Gurusingham Sitta Sittampalam
- The Institute for Advancing Medical Innovation, The University of Kansas Medical Center, Kansas City, Kansas
- Department of Pharmacology, Toxicology, and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
16
|
Elliott NT, Yuan F. A review of three-dimensional in vitro tissue models for drug discovery and transport studies. J Pharm Sci 2010; 100:59-74. [PMID: 20533556 DOI: 10.1002/jps.22257] [Citation(s) in RCA: 318] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 05/04/2010] [Indexed: 12/12/2022]
Abstract
The use of animal models in drug discovery studies presents issues with feasibility and ethical concerns. To address these limitations, in vitro tissue models have been developed to provide a means for systematic, repetitive, and quantitative investigation of drugs. By eliminating or reducing the need for animal subjects, these models can serve as platforms for more tightly controlled, high-throughput screening of drugs and for pharmacokinetic and pharmacodynamic analyses of drugs. The focus of this review is three-dimensional (3D) tissue models that can capture cell-cell and cell-matrix interactions. Compared to the 2D culture of cell monolayers, 3D models more closely mimic native tissues since the cellular microenvironment established in the 3D models often plays a significant role in disease progression and cellular responses to drugs. A growing body of research has been published in the literature, which highlights the benefits of the 3D in vitro models of various tissues. This review provides an overview of some successful 3D in vitro models that have been developed to mimic liver, breast, cardiac, muscle, bone, and corneal tissues as well as malignant tissues in solid tumors.
Collapse
Affiliation(s)
- Nelita T Elliott
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, PO Box 90281, Durham, North Carolina 27708, USA
| | | |
Collapse
|
17
|
Hummitzsch K, Ricken AM, Kloss D, Erdmann S, Nowicki MS, Rothermel A, Robitzki AA, Spanel-Borowski K. Spheroids of granulosa cells provide an in vitro model for programmed cell death coupled to steroidogenesis. Differentiation 2008; 77:60-9. [PMID: 19281765 DOI: 10.1016/j.diff.2008.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2008] [Revised: 06/06/2008] [Accepted: 06/12/2008] [Indexed: 12/19/2022]
Abstract
UNLABELLED We describe the use of rotary cultures (72 rpm) as an excellent method for generating spheroids from dispersed bovine granulosa cells (GC). The GC spheroids were symmetrical (diameter between 100 and 200 microm), easily accessible, and could be obtained at high yields. On day one, the spheroids showed a two-layered outer zone of cells that stained lighter than the inner zone in semi-thin sections. Bromodeoxyuridine (BrdU) uptake was frequent and randomly distributed. By day two, a striking decrease in BrdU uptake was noted. Apoptotic bodies appeared up to day four, as did TUNEL and propidium iodide labelled dead cells. At that time, the inner zone contained cells with large-sized vacuoles and the core was amorphous. The large-sized vacuoles were identified at the ultrastructural level and represented autophagosomes and autophagolysosomes that were in different stages of development. Surprisingly, conspicuous signs of cell death were accompanied by an increase in spontaneous luteinization compared to conventional stationary cultures. We detected high levels of progesterone (immunoassay) accompanied by high levels of the proteins and enzymes relevant for steroidogenesis (StAR, P450scc, 3beta-HSD by immunoblot and immunohistochemistry, respectively). CONCLUSIONS Concomitant to cell death, GC spheroids augment progesterone synthesis. The GC spheroids provide an ideal model for studying steroidogenesis coupled to programmed cell death at the level of the mitochondria.
Collapse
Affiliation(s)
- Katja Hummitzsch
- Department of Anatomy, University of Leipzig, D-04103 Leipzig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Pierzchalski A, Robitzki A, Mittag A, Emmrich F, Sack U, O'Connor JE, Bocsi J, Tárnok A. Cytomics and nanobioengineering. CYTOMETRY PART B-CLINICAL CYTOMETRY 2008; 74:416-26. [PMID: 18814265 DOI: 10.1002/cyto.b.20453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The finding that an individual's genome differs as much as by many million variants from that of the human reference assembly diminished the great enthusiasm that every disease could be predicted based on nucleotide polymorphisms. Even individual cells of an organ may be specifically equipped to perform specific tasks and that the information of individual cells in a cell system is key information to understand function or dysfunction. Therefore, cytomics received great attention during the last years as it allows to quantitatively and qualitatively analyzing great number of individual cells, cell constituents, and of their intracellular and functional interactions in a cellular system and also giving the concept of analysis of these data.Exhaustive data extraction from multiparametric assays and multiple tests are the prerequisite for prediction of drug toxicity. Cytomics, as novel approach for unsupervised data analysis give a chance to find the most predictive parameters, which describe best the toxicity of a chemical. Cytomics is intrinsically connected to drug development and drug discovery.Focused on small structures, nanobioengineering is the ideal partner of cytomics, the systems biological discipline for cell population analysis. Realizing the idea "from the molecule to the patient" develops and offers chemical compounds, proteins, and other biomolecules, cells as well as tissues as instruments and products for a wide variety of biotechnological and biomedical applications.The integrative nanobioengineering combining different disciplines of nanotechnology will promote the development of innovative therapies and diagnostic methods. It can improve the precision of the measurements with focus on single cell analysis. By nanobioengineering and whole body imaging techniques, cytomics covers the field from molecules through bacterial cells, eukaryotic tissues, and organs to small animal live analysis. Toxicological testing and medical drug development are currently strongly broadening. It harbors the promise to substantially impact on various fields of biomedicine, drug discovery, and predictive medicine.As the number of scientific data is rising exponentially, new data analysis tools and strategies like cytomics and nanobioengineering take a lead and get closer to application. Bionanoengineering may strongly support the quantitative data supply, thus strengthening the rational for cytomics approach.
Collapse
Affiliation(s)
- Arkadiusz Pierzchalski
- Department of Pediatric Cardiology, Heart Center Leipzig, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Kloss D, Fischer M, Rothermel A, Simon JC, Robitzki AA. Drug testing on 3D in vitro tissues trapped on a microcavity chip. LAB ON A CHIP 2008; 8:879-884. [PMID: 18497906 DOI: 10.1039/b800394g] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Close to realistic responses to anti-cancer drugs are not adequately provided in monolayer or single cells assays. 3-dimensional multicellular cultures (spheroids) mimicking in vivo-like conditions are established as cell biological models for microtumors/metastases. For a non-invasive real-time monitoring of the electrical parameters of such spheroid cultures we designed, fabricated and tested a 3D multifunctional electrode-based microcavity array. In a non-adherent assay acute tests with tumor spheroids were done maintaining their spherical shape and cellular arrangement. The sensor chip with 15 individual square microcavities containing four gold electrodes each was used for impedance spectroscopy to analyze the tissue models in terms of morphological and structural changes. Cell type specific differences in the spectra and varying responses to several anti-tumor drugs were found. Further development of the prototype will provide a promising tool for the use in pharmacological high-throughput studies.
Collapse
Affiliation(s)
- Daniel Kloss
- Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Division of Molecular Biological-Biochemical Processing Technology, Deutscher Platz 5, 04103, Leipzig, Germany
| | | | | | | | | |
Collapse
|