1
|
Kito K, Hayashi M, Kaneko T. Electrophysiological analysis of hyperkalemic cardiomyocytes using a multielectrode array system. Biophys Physicobiol 2024; 21:e210026. [PMID: 39963594 PMCID: PMC11832246 DOI: 10.2142/biophysico.bppb-v21.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/19/2024] [Indexed: 02/20/2025] Open
Abstract
The action potential of cardiomyocytes is controlled by electrolytes in serum such as Na+, K+ and Ca2+. Hyperkalemia, which refers to an abnormally high concentration of K+ in the blood, can induce lethal arrythmia. In this study, the extracellular potentials on a sheet of chick embryonic cardiomyocytes were investigated at increasing K+ concentrations using a multielectrode array system. We observed that the interspike interval (ISI) was prolonged by approximately 3.5 times; dV/dt (the slope of a waveform) was decreased by more than five times; the field potential duration (FPD) was shortened by 20%, and the conduction velocity was about half at 12 mM K+ against the control (4 mM K+). In calcium therapy for hyperkalemia, although the prolongation of ISI under hyperkalemic conditions was restored, the slowing of conduction velocity, the decrease in dV/dt, and the shortening of FPD were not recovered by increasing the extracellular Ca2+ concentration. These findings provide a comprehensive understanding of cardiomyocytes in hyperkalemic conditions. Electrophysiological analysis by varying the extracellular concentrations of multiple types of electrolytes will be useful for the further discussion of the results of this study and for the interpretation of the waveforms obtained by measuring the extracellular potential.
Collapse
Affiliation(s)
- Kentaro Kito
- Department of Frontier Bioscience, Graduate School of Science & Engineering, Hosei University, Koganei, Tokyo 184-8584, Japan
| | - Masahito Hayashi
- Department of Frontier Bioscience, Graduate School of Science & Engineering, Hosei University, Koganei, Tokyo 184-8584, Japan
| | - Tomoyuki Kaneko
- Department of Frontier Bioscience, Graduate School of Science & Engineering, Hosei University, Koganei, Tokyo 184-8584, Japan
| |
Collapse
|
2
|
Murphy JF, Costa KD, Turnbull IC. Rianú: Multi-tissue tracking software for increased throughput of engineered cardiac tissue screening. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE UPDATE 2023; 3:100107. [PMID: 37476002 PMCID: PMC10359020 DOI: 10.1016/j.cmpbup.2023.100107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Background The field of tissue engineering has provided valuable three-dimensional species-specific models of the human myocardium in the form of human Engineered Cardiac Tissues (hECTs) and similar constructs. However, hECT systems are often bottlenecked by a lack of openly available software that can collect data from multiple tissues at a time, even in multi-tissue bioreactors, which limits throughput in phenotypic and therapeutic screening applications. Methods We developed Rianú, an open-source web application capable of simultaneously tracking multiple hECTs on flexible end-posts. This software is operating system agnostic and deployable on a remote server, accessible via a web browser with no local hardware or software requirements. The software incorporates object-tracking capabilities for multiple objects simultaneously, an algorithm for twitch tracing analysis and contractile force calculation, and a data compilation system for comparative analysis within and amongst groups. Validation tests were performed using in-silico and in-vitro experiments for comparison with established methods and interventions. Results Rianú was able to detect the displacement of the flexible end-posts with a sub-pixel sensitivity of 0.555 px/post (minimum increment in post displacement) and a lower limit of 1.665 px/post (minimum post displacement). Compared to our established reference for contractility assessment, Rianú had a high correlation for all parameters analyzed (ranging from R 2 = 0.7514 to R 2 = 0.9695 ), demonstrating its high accuracy and reliability. Conclusions Rianú provides simultaneous tracking of multiple hECTs, expediting the recording and analysis processes, and simplifies time-based intervention studies. It also allows data collection from different formats and has scale-up capabilities proportional to the number of tissues per field of view. These capabilities will enhance throughput of hECTs and similar assays for in-vitro analysis in disease modeling and drug screening applications.
Collapse
Affiliation(s)
- Jack F. Murphy
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| | - Kevin D. Costa
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| | - Irene C. Turnbull
- Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1014, New York City, 10029, NY, USA
| |
Collapse
|
3
|
Sakamoto K, Matsumoto S, Abe N, Sentoku M, Yasuda K. Importance of Spatial Arrangement of Cardiomyocyte Network for Precise and Stable On-Chip Predictive Cardiotoxicity Measurement. MICROMACHINES 2023; 14:854. [PMID: 37421087 DOI: 10.3390/mi14040854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 07/09/2023]
Abstract
One of the advantages of human stem cell-derived cell-based preclinical screening is the reduction of the false negative/positive misjudgment of lead compounds for predicting their effectiveness and risks during the early stage of development. However, as the community effect of cells was neglected in the conventional single cell-based in vitro screening, the potential difference in results caused by the cell number and their spatial arrangement differences has not yet been sufficiently evaluated. Here, we have investigated the effect of the community size and spatial arrangement difference for cardiomyocyte network response against the proarrhythmic compounds from the viewpoint of in vitro cardiotoxicity. Using three different typical types of cell networks of cardiomyocytes, small cluster, large square sheet, and large closed-loop sheet were formed in shaped agarose microchambers fabricated on a multielectrode array chip simultaneously, and their responses were compared against the proarrhythmic compound, E-4031. The interspike intervals (ISIs) in large square sheets and closed-loop sheets were durable and maintained stable against E-4031 even at a high dose of 100 nM. In contrast, those in the small cluster, which fluctuated even without E-4031, acquired stable beating reflecting the antiarrhythmic efficacy of E-4031 from a 10 nM medium dose administration. The repolarization index, field potential duration (FPD), was prolonged in closed-loop sheets with 10 nM E-4031, even though small clusters and large sheets remained normal at this concentration. Moreover, FPDs of large sheets were the most durable against E-4031 among the three geometries of cardiomyocyte networks. The results showed the apparent spatial arrangement dependence on the stability of their interspike intervals, and FPD prolongation, indicating the importance of the geometry control of cell networks for representing the appropriate response of cardiomyocytes against the adequate amount of compounds for in vitro ion channel measurement.
Collapse
Affiliation(s)
- Kazufumi Sakamoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Suguru Matsumoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Nanami Abe
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Mitsuru Sentoku
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo 169-8555, Japan
| |
Collapse
|
4
|
Bersell KR, Yang T, Mosley JD, Glazer AM, Hale AT, Kryshtal DO, Kim K, Steimle JD, Brown JD, Salem JE, Campbell CC, Hong CC, Wells QS, Johnson AN, Short L, Blair MA, Behr ER, Petropoulou E, Jamshidi Y, Benson MD, Keyes MJ, Ngo D, Vasan RS, Yang Q, Gerszten RE, Shaffer C, Parikh S, Sheng Q, Kannankeril PJ, Moskowitz IP, York JD, Wang TJ, Knollmann BC, Roden DM. Transcriptional Dysregulation Underlies Both Monogenic Arrhythmia Syndrome and Common Modifiers of Cardiac Repolarization. Circulation 2023; 147:824-840. [PMID: 36524479 PMCID: PMC9992308 DOI: 10.1161/circulationaha.122.062193] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Brugada syndrome (BrS) is an inherited arrhythmia syndrome caused by loss-of-function variants in the cardiac sodium channel gene SCN5A (sodium voltage-gated channel alpha subunit 5) in ≈20% of subjects. We identified a family with 4 individuals diagnosed with BrS harboring the rare G145R missense variant in the cardiac transcription factor TBX5 (T-box transcription factor 5) and no SCN5A variant. METHODS We generated induced pluripotent stem cells (iPSCs) from 2 members of a family carrying TBX5-G145R and diagnosed with Brugada syndrome. After differentiation to iPSC-derived cardiomyocytes (iPSC-CMs), electrophysiologic characteristics were assessed by voltage- and current-clamp experiments (n=9 to 21 cells per group) and transcriptional differences by RNA sequencing (n=3 samples per group), and compared with iPSC-CMs in which G145R was corrected by CRISPR/Cas9 approaches. The role of platelet-derived growth factor (PDGF)/phosphoinositide 3-kinase (PI3K) pathway was elucidated by small molecule perturbation. The rate-corrected QT (QTc) interval association with serum PDGF was tested in the Framingham Heart Study cohort (n=1893 individuals). RESULTS TBX5-G145R reduced transcriptional activity and caused multiple electrophysiologic abnormalities, including decreased peak and enhanced "late" cardiac sodium current (INa), which were entirely corrected by editing G145R to wild-type. Transcriptional profiling and functional assays in genome-unedited and -edited iPSC-CMs showed direct SCN5A down-regulation caused decreased peak INa, and that reduced PDGF receptor (PDGFRA [platelet-derived growth factor receptor α]) expression and blunted signal transduction to PI3K was implicated in enhanced late INa. Tbx5 regulation of the PDGF axis increased arrhythmia risk due to disruption of PDGF signaling and was conserved in murine model systems. PDGF receptor blockade markedly prolonged normal iPSC-CM action potentials and plasma levels of PDGF in the Framingham Heart Study were inversely correlated with the QTc interval (P<0.001). CONCLUSIONS These results not only establish decreased SCN5A transcription by the TBX5 variant as a cause of BrS, but also reveal a new general transcriptional mechanism of arrhythmogenesis of enhanced late sodium current caused by reduced PDGF receptor-mediated PI3K signaling.
Collapse
Affiliation(s)
- Kevin R Bersell
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Tao Yang
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Jonathan D Mosley
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Andrew M Glazer
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Andrew T Hale
- Biochemistry (A.T.H., J.D.Y.), Vanderbilt University, Nashville, TN
| | - Dmytro O Kryshtal
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Kyungsoo Kim
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Jeffrey D Steimle
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, IL (J.D.S., I.P.M.)
| | - Jonathan D Brown
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Joe-Elie Salem
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, CIC-1901, Sorbonne University, Paris, France (J-E.S.)
- Sorbonne Universités, UPMC Univ Paris 06, Faculty of Medicine, France (J-E.S.)
| | - Courtney C Campbell
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Charles C Hong
- Department of Medicine, University of Maryland School of Medicine, Baltimore (C.C.H.)
| | - Quinn S Wells
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Biomedical Informatics (Q.S.W., D.M.R.), Vanderbilt University, Nashville, TN
| | - Amanda N Johnson
- Molecular Physiology and Biophysics (A.N.J.), Vanderbilt University, Nashville, TN
| | - Laura Short
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Marcia A Blair
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | | | - Evmorfia Petropoulou
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Institute, St George's, University of London and St George's University Hospitals National Health Service Foundation Trust, London, UK (E.P., Y.J.)
| | - Yalda Jamshidi
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Institute, St George's, University of London and St George's University Hospitals National Health Service Foundation Trust, London, UK (E.P., Y.J.)
| | - Mark D Benson
- Cardiovascular Research Center (E.J.B., M.D.B., M.J.K., R.E.G.), Beth Israel Deaconess Hospital, Boston, MA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA (M.D.B.)
| | - Michelle J Keyes
- Cardiovascular Research Center (E.J.B., M.D.B., M.J.K., R.E.G.), Beth Israel Deaconess Hospital, Boston, MA
| | - Debby Ngo
- Division of Pulmonary and Cardiovascular Medicine (D.N., R.E.G.), Beth Israel Deaconess Hospital, Boston, MA
| | | | - Qiong Yang
- Boston University School of Medicine, MA (R.S.V., Q.Y.)
| | - Robert E Gerszten
- Cardiovascular Research Center (E.J.B., M.D.B., M.J.K., R.E.G.), Beth Israel Deaconess Hospital, Boston, MA
- Division of Pulmonary and Cardiovascular Medicine (D.N., R.E.G.), Beth Israel Deaconess Hospital, Boston, MA
| | - Christian Shaffer
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Shan Parikh
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | | | | | - Ivan P Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, IL (J.D.S., I.P.M.)
| | - John D York
- Biochemistry (A.T.H., J.D.Y.), Vanderbilt University, Nashville, TN
| | - Thomas J Wang
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Bjorn C Knollmann
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
| | - Dan M Roden
- Departments of Pharmacology (K.R.B., A.M.G., D.O.K., K.K., J-E.S., C.C.C., Q.S.W., S.P., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Medicine (T.Y., J.D.M., J.D.B., J-E.S., Q.S.W., L.S., M.A.B., C.S., T.J.W., B.C.K., D.M.R.), Vanderbilt University, Nashville, TN
- Biomedical Informatics (Q.S.W., D.M.R.), Vanderbilt University, Nashville, TN
| |
Collapse
|
5
|
Sentoku M, Hashimoto H, Iida K, Endo M, Yasuda K. Photothermal Agarose Microfabrication Technology for Collective Cell Migration Analysis. MICROMACHINES 2021; 12:1015. [PMID: 34577661 PMCID: PMC8467839 DOI: 10.3390/mi12091015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/28/2022]
Abstract
Agarose photothermal microfabrication technology is one of the micropatterning techniques that has the advantage of simple and flexible real-time fabrication even during the cultivation of cells. To examine the ability and limitation of the agarose microstructures, we investigated the collective epithelial cell migration behavior in two-dimensional agarose confined structures. Agarose microchannels from 10 to 211 micrometer width were fabricated with a spot heating of a focused 1480 nm wavelength infrared laser to the thin agarose layer coated on the cultivation dish after the cells occupied the reservoir. The collective cell migration velocity maintained constant regardless of their extension distance, whereas the width dependency of those velocities was maximized around 30 micrometer width and decreased both in the narrower and wider microchannels. The single-cell tracking revealed that the decrease of velocity in the narrower width was caused by the apparent increase of aspect ratio of cell shape (up to 8.9). In contrast, the decrease in the wider channels was mainly caused by the increase of the random walk-like behavior of component cells. The results confirmed the advantages of this method: (1) flexible fabrication without any pre-designing, (2) modification even during cultivation, and (3) the cells were confined in the agarose geometry.
Collapse
Affiliation(s)
- Mitsuru Sentoku
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (M.S.); (H.H.); (K.I.)
| | - Hiromichi Hashimoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (M.S.); (H.H.); (K.I.)
| | - Kento Iida
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (M.S.); (H.H.); (K.I.)
| | - Masaharu Endo
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan;
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (M.S.); (H.H.); (K.I.)
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan;
| |
Collapse
|
6
|
Tanaka Y, Watanabe H, Shimoda K, Sakamoto K, Hondo Y, Sentoku M, Sekine R, Kikuchi T, Yasuda K. Stepwise neuronal network pattern formation in agarose gel during cultivation using non-destructive microneedle photothermal microfabrication. Sci Rep 2021; 11:14656. [PMID: 34282174 PMCID: PMC8289850 DOI: 10.1038/s41598-021-93988-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/05/2021] [Indexed: 01/25/2023] Open
Abstract
Conventional neuronal network pattern formation techniques cannot control the arrangement of axons and dendrites because network structures must be fixed before neurite differentiation. To overcome this limitation, we developed a non-destructive stepwise microfabrication technique that can be used to alter microchannels within agarose to guide neurites during elongation. Micropatterns were formed in thin agarose layer coating of a cultivation dish using the tip of a 0.7 [Formula: see text]-diameter platinum-coated glass microneedle heated by a focused 1064-nm wavelength infrared laser, which has no absorbance of water. As the size of the heat source was 0.7 [Formula: see text], which is smaller than the laser wavelength, the temperature fell to 45 [Formula: see text] within a distance of 7.0 [Formula: see text] from the edge of the etched agarose microchannel. We exploited the fast temperature decay property to guide cell-to-cell connection during neuronal network cultivation. The first neurite of a hippocampal cell from a microchamber was guided to a microchannel leading to the target neuron with stepwise etching of the micrometer resolution microchannel in the agarose layer, and the elongated neurites were not damaged by the heat of etching. The results indicate the potential of this new technique for fully direction-controlled on-chip neuronal network studies.
Collapse
Affiliation(s)
- Yuhei Tanaka
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Haruki Watanabe
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Kenji Shimoda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Kazufumi Sakamoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Yoshitsune Hondo
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Mitsuru Sentoku
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Rikuto Sekine
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Takahito Kikuchi
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan.
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Tokyo, 169-8555, Japan.
| |
Collapse
|
7
|
Yu J, Cai P, Chen X. Structural Regulation of Myocytes in Engineered Healthy and Diseased Cardiac Models. ACS APPLIED BIO MATERIALS 2021; 4:267-276. [DOI: 10.1021/acsabm.0c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Yu
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Pingqiang Cai
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Xiaodong Chen
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
8
|
Sakamoto K, Aoki S, Tanaka Y, Shimoda K, Hondo Y, Yasuda K. Geometric Understanding of Local Fluctuation Distribution of Conduction Time in Lined-Up Cardiomyocyte Network in Agarose-Microfabrication Multi-Electrode Measurement Assay. MICROMACHINES 2020; 11:mi11121105. [PMID: 33327568 PMCID: PMC7765075 DOI: 10.3390/mi11121105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 11/17/2022]
Abstract
We examined characteristics of the propagation of conduction in width-controlled cardiomyocyte cell networks for understanding the contribution of the geometrical arrangement of cardiomyocytes for their local fluctuation distribution. We tracked a series of extracellular field potentials of linearly lined-up human embryonic stem (ES) cell-derived cardiomyocytes and mouse primary cardiomyocytes with 100 kHz sampling intervals of multi-electrodes signal acquisitions and an agarose microfabrication technology to localize the cardiomyocyte geometries in the lined-up cell networks with 100–300 μm wide agarose microstructures. Conduction time between two neighbor microelectrodes (300 μm) showed Gaussian distribution. However, the distributions maintained their form regardless of its propagation distances up to 1.5 mm, meaning propagation diffusion did not occur. In contrast, when Quinidine was applied, the propagation time distributions were increased as the faster firing regulation simulation predicted. The results indicate the “faster firing regulation” is not sufficient to explain the conservation of the propagation time distribution in cardiomyocyte networks but should be expanded with a kind of community effect of cell networks, such as the lower fluctuation regulation.
Collapse
Affiliation(s)
- Kazufumi Sakamoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
| | - Shota Aoki
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
| | - Yuhei Tanaka
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
| | - Kenji Shimoda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
| | - Yoshitsune Hondo
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan; (K.S.); (S.A.); (Y.T.); (K.S.); (Y.H.)
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Tokyo 169-8555, Japan
- Correspondence:
| |
Collapse
|
9
|
Wei X, Zhuang L, Li H, He C, Wan H, Hu N, Wang P. Advances in Multidimensional Cardiac Biosensing Technologies: From Electrophysiology to Mechanical Motion and Contractile Force. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2005828. [PMID: 33230867 DOI: 10.1002/smll.202005828] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is currently a leading killer to human, while drug-induced cardiotoxicity remains the main cause of the withdrawal and attrition of drugs. Taking clinical correlation and throughput into account, cardiomyocyte is perfect as in vitro cardiac model for heart disease modeling, drug discovery, and cardiotoxicity assessment by accurately measuring the physiological multiparameters of cardiomyocytes. Remarkably, cardiomyocytes present both electrophysiological and biomechanical characteristics due to the unique excitation-contraction coupling, which plays a significant role in studying the cardiomyocytes. This review mainly focuses on the recent advances of biosensing technologies for the 2D and 3D cardiac models with three special properties: electrophysiology, mechanical motion, and contractile force. These high-performance multidimensional cardiac models are popular and effective to rebuild and mimic the heart in vitro. To help understand the high-quality and accurate physiologies, related detection techniques are highly demanded, from microtechnology to nanotechnology, from extracellular to intracellular recording, from multiple cells to single cell, and from planar to 3D models. Furthermore, the characteristics, advantages, limitations, and applications of these cardiac biosensing technologies, as well as the future development prospects should contribute to the systematization and expansion of knowledge.
Collapse
Affiliation(s)
- Xinwei Wei
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Liujing Zhuang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Hongbo Li
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chuanjiang He
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
| | - Hao Wan
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and Technologies, Guangdong Province Key Laboratory of Display Material and Technology, School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ping Wang
- Department of Biomedical Engineering, Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
10
|
Arai S, Lloyd K, Takahashi T, Mammoto K, Miyazawa T, Tamura K, Kaneko T, Ishida K, Moriyama Y, Mitsui T. Dynamic Properties of Heart Fragments from Different Regions and Their Synchronization. Bioengineering (Basel) 2020; 7:E81. [PMID: 32751255 PMCID: PMC7552607 DOI: 10.3390/bioengineering7030081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/29/2022] Open
Abstract
The dynamic properties of the heart differ based on the regions that effectively circulate blood throughout the body with each heartbeat. These properties, including the inter-beat interval (IBI) of autonomous beat activity, are retained even in in vitro tissue fragments. However, details of beat dynamics have not been well analyzed, particularly at the sub-mm scale, although such dynamics of size are important for regenerative medicine and computational studies of the heart. We analyzed the beat dynamics in sub-mm tissue fragments from atria and ventricles of hearts obtained from chick embryos over a period of 40 h. The IBI and contraction speed differed by region and atrial fragments retained their values for a longer time. The major finding of this study is synchronization of these fragment pairs physically attached to each other. The probability of achieving this and the time required differ for regional pairs: atrium-atrium, ventricle-ventricle, or atrium-ventricle. Furthermore, the time required to achieve 1:1 synchronization does not depend on the proximity of initial IBI of paired fragments. Various interesting phenomena, such as 1:n synchronization and a reentrant-like beat sequence, are revealed during synchronization. Finally, our observation of fragment dynamics indicates that mechanical motion itself contributes to the synchronization of atria.
Collapse
Affiliation(s)
- Shin Arai
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kento Lloyd
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Tomonori Takahashi
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kazuki Mammoto
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Takashi Miyazawa
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kei Tamura
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Tomoyuki Kaneko
- Department of Frontier Bioscience, Hosei University, Koganei, Tokyo 184-8584, Japan;
| | - Kentaro Ishida
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Yuuta Moriyama
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Toshiyuki Mitsui
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| |
Collapse
|
11
|
Dominant rule of community effect in synchronized beating behavior of cardiomyocyte networks. Biophys Rev 2020; 12:481-501. [PMID: 32367300 DOI: 10.1007/s12551-020-00688-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/03/2020] [Indexed: 10/24/2022] Open
Abstract
Exploiting the combination of latest microfabrication technologies and single cell measurement technologies, we can measure the interactions of single cells, and cell networks from "algebraic" and "geometric" perspectives under the full control of their environments and interactions. However, the experimental constructive single cell-based approach still remains the limitations regarding the quality and condition control of those cells. To overcome these limitations, mathematical modeling is one of the most powerful complementary approaches. In this review, we first explain our on-chip experimental methods for constructive approach, and we introduce the results of the "community effect" of beating cardiomyocyte networks as an example of this approach. On-chip analysis revealed that (1) synchronized interbeat intervals (IBIs) of cell networks were followed to the more stable beating cells even their IBIs were slower than the other cells, which is against the conventional faster firing regulation or "overdrive suppression," and (2) fluctuation of IBIs of cardiomyocyte networks decreased according to the increase of the number of connected cells regardless of their geometry. The mathematical simulation of this synchronous behavior of cardiomyocyte networks also fitted well with the experimental results after incorporating the fluctuation-dissipation theorem into the oscillating stochastic phase model, in which the concept of spatially arranged cardiomyocyte networks was involved. The constructive experiments and mathematical modeling indicated the dominant rule of synchronization behavior of beating cardiomyocyte networks is a kind of stability-oriented synchronization phenomenon as the "community effect" or a fluctuation-dissipation phenomenon. Finally, as a practical application of this approach, the predictive cardiotoxicity is introduced.
Collapse
|
12
|
Takano M, Yura K, Uyeda T, Yasuda K. Biophysics at Waseda University. Biophys Rev 2020; 12:225-232. [PMID: 32157615 PMCID: PMC7242523 DOI: 10.1007/s12551-020-00638-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/20/2022] Open
Abstract
Biophysics in Waseda University was started in 1965 as one of the three key research areas that constitute the Physics Department. In the biophysics group, one theoretical lab and two experimental labs are now working on the cutting-edge themes on biophysics, disseminating the ideas and knowledge of biophysics to undergraduate and graduate students from the viewpoint of physics.
Collapse
Affiliation(s)
- Mitsunori Takano
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kei Yura
- Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Taro Uyeda
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Kenji Yasuda
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
- Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.
| |
Collapse
|
13
|
|
14
|
Modeling of LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Cells 2019; 8:cells8060594. [PMID: 31208058 PMCID: PMC6627421 DOI: 10.3390/cells8060594] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the leading causes of heart failure and heart transplantation. A portion of familial DCM is due to mutations in the LMNA gene encoding the nuclear lamina proteins lamin A and C and without adequate treatment these patients have a poor prognosis. To get better insights into pathobiology behind this disease, we focused on modeling LMNA-related DCM using human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM). Primary skin fibroblasts from DCM patients carrying the most prevalent Finnish founder mutation (p.S143P) in LMNA were reprogrammed into hiPSCs and further differentiated into cardiomyocytes (CMs). The cellular structure, functionality as well as gene and protein expression were assessed in detail. While mutant hiPSC-CMs presented virtually normal sarcomere structure under normoxia, dramatic sarcomere damage and an increased sensitivity to cellular stress was observed after hypoxia. A detailed electrophysiological evaluation revealed bradyarrhythmia and increased occurrence of arrhythmias in mutant hiPSC-CMs on β-adrenergic stimulation. Mutant hiPSC-CMs also showed increased sensitivity to hypoxia on microelectrode array and altered Ca2+ dynamics. Taken together, p.S143P hiPSC-CM model mimics hallmarks of LMNA-related DCM and provides a useful tool to study the underlying cellular mechanisms of accelerated cardiac degeneration in this disease.
Collapse
|
15
|
Asahi Y, Nomura F, Abe Y, Doi M, Sakakura T, Takasuna K, Yasuda K. Electrophysiological evaluation of pentamidine and 17-AAG in human stem cell-derived cardiomyocytes for safety assessment. Eur J Pharmacol 2019; 842:221-230. [DOI: 10.1016/j.ejphar.2018.10.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 10/28/2022]
|
16
|
Asahi Y, Hamada T, Hattori A, Matsuura K, Odaka M, Nomura F, Kaneko T, Abe Y, Takasuna K, Sanbuissho A, Yasuda K. On-chip spatiotemporal electrophysiological analysis of human stem cell derived cardiomyocytes enables quantitative assessment of proarrhythmia in drug development. Sci Rep 2018; 8:14536. [PMID: 30266924 PMCID: PMC6162288 DOI: 10.1038/s41598-018-32921-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 09/17/2018] [Indexed: 12/17/2022] Open
Abstract
We examined a simultaneous combined spatiotemporal field potential duration (FPD) and cell-to-cell conduction time (CT) in lined-up shaped human embryonic stem cell-derived cardiomyocytes (hESC-CMs) using an on-chip multielectrode array (MEA) system to evaluate two origins of lethal arrhythmia, repolarization and depolarization. The repolarization index, FPD, was prolonged by E-4031 and astemizole, and shortened by verapamil, flecainide and terfenadine at 10 times higher than therapeutic plasma concentrations of each drug, but it did not change after lidocaine treatment up to 100 μM. CT was increased by astemizol, flecainide, terfenadine, and lidocaine at equivalent concentrations of Nav1.5 IC50, suggesting that CT may be an index of cardiac depolarization because the increase in CT (i.e., decrease in cell-to-cell conduction speed) was relevant to Nav1.5 inhibition. Fluctuations (short-term variability; STV) of FPD and CT, STVFPD and STVCT also discriminated between torsadogenic and non-torsadogenic compounds with significant increases in their fluctuation values, enabling precise prediction of arrhythmogenic risk as potential new indices.
Collapse
Affiliation(s)
- Yumiko Asahi
- Medicinal Safety Research Laboratories, Kasai R&D Center, Daiichi-Sankyo Co. Ltd., Edogawa, Tokyo, 134-8630, Japan
| | - Tomoyo Hamada
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
- Chiome Bioscience Inc. Shibuya, Tokyo, 151-0071, Japan
| | - Akihiro Hattori
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
- Organization for University Research Initiatives, Waseda University, 3-14-9 Ookubo, Shinjuku, Tokyo, 169-0072, Japan
- Waseda Bioscience Research Institute in Singapore (WABOIS), Helios, 11 Biopolis Way, 138667, Singapore
| | - Kenji Matsuura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
- Organization for University Research Initiatives, Waseda University, 3-14-9 Ookubo, Shinjuku, Tokyo, 169-0072, Japan
- Waseda Bioscience Research Institute in Singapore (WABOIS), Helios, 11 Biopolis Way, 138667, Singapore
| | - Masao Odaka
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
- Organization for University Research Initiatives, Waseda University, 3-14-9 Ookubo, Shinjuku, Tokyo, 169-0072, Japan
- Waseda Bioscience Research Institute in Singapore (WABOIS), Helios, 11 Biopolis Way, 138667, Singapore
| | - Fumimasa Nomura
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
| | - Tomoyuki Kaneko
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan
- Department of Frontier Bioscience, Hosei Univ., Koganei, Tokyo, 184-8584, Japan
| | - Yasuyuki Abe
- Medicinal Safety Research Laboratories, Kasai R&D Center, Daiichi-Sankyo Co. Ltd., Edogawa, Tokyo, 134-8630, Japan
| | - Kiyoshi Takasuna
- Medicinal Safety Research Laboratories, Kasai R&D Center, Daiichi-Sankyo Co. Ltd., Edogawa, Tokyo, 134-8630, Japan
| | - Atsushi Sanbuissho
- Medicinal Safety Research Laboratories, Kasai R&D Center, Daiichi-Sankyo Co. Ltd., Edogawa, Tokyo, 134-8630, Japan
| | - Kenji Yasuda
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo, 101-0062, Japan.
- Organization for University Research Initiatives, Waseda University, 3-14-9 Ookubo, Shinjuku, Tokyo, 169-0072, Japan.
- Waseda Bioscience Research Institute in Singapore (WABOIS), Helios, 11 Biopolis Way, 138667, Singapore.
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo, 169-8555, Japan.
- Department of Physics, School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku, Tokyo, 169-8555, Japan.
| |
Collapse
|
17
|
Oleaga C, Riu A, Rothemund S, Lavado A, McAleer CW, Long CJ, Persaud K, Narasimhan NS, Tran M, Roles J, Carmona-Moran CA, Sasserath T, Elbrecht DH, Kumanchik L, Bridges LR, Martin C, Schnepper MT, Ekman G, Jackson M, Wang YI, Note R, Langer J, Teissier S, Hickman JJ. Investigation of the effect of hepatic metabolism on off-target cardiotoxicity in a multi-organ human-on-a-chip system. Biomaterials 2018; 182:176-190. [PMID: 30130706 DOI: 10.1016/j.biomaterials.2018.07.062] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022]
Abstract
Regulation of cosmetic testing and poor predictivity of preclinical drug studies has spurred efforts to develop new methods for systemic toxicity. Current in vitro assays do not fully represent physiology, often lacking xenobiotic metabolism. Functional human multi-organ systems containing iPSC derived cardiomyocytes and primary hepatocytes were maintained under flow using a low-volume pumpless system in a serum-free medium. The functional readouts for contractile force and electrical conductivity enabled the non-invasive study of cardiac function. The presence of the hepatocytes in the system induced cardiotoxic effects from cyclophosphamide and reduced them for terfenadine due to drug metabolism, as expected from each compound's pharmacology. A computational fluid dynamics simulation enabled the prediction of terfenadine-fexofenadine pharmacokinetics, which was validated by HPLC-MS. This in vitro platform recapitulates primary aspects of the in vivo crosstalk between heart and liver and enables pharmacological studies, involving both organs in a single in vitro platform. The system enables non-invasive readouts of cardiotoxicity of drugs and their metabolites. Hepatotoxicity can also be evaluated by biomarker analysis and change in metabolic function. Integration of metabolic function in toxicology models can improve adverse effects prediction in preclinical studies and this system could also be used for chronic studies as well.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Anne Riu
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - Sandra Rothemund
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Andrea Lavado
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Christopher W McAleer
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Keisha Persaud
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | | | - My Tran
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Jeffry Roles
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Carlos A Carmona-Moran
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Trevor Sasserath
- Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Daniel H Elbrecht
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Hesperos, Inc., 3259 Progress Dr, Room 158, Orlando, FL 32826, USA
| | - Lee Kumanchik
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | | | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Mark T Schnepper
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Max Jackson
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA
| | - Ying I Wang
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Reine Note
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - Jessica Langer
- L'Oreal Research, and Innovation Division, Clark, NJ, USA
| | - Silvia Teissier
- L'Oreal Research, and Innovation Division, Aulnay-sous-Bois, France
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32826, USA.
| |
Collapse
|
18
|
Mandenius CF. Conceptual Design of Micro-Bioreactors and Organ-on-Chips for Studies of Cell Cultures. Bioengineering (Basel) 2018; 5:E56. [PMID: 30029542 PMCID: PMC6164921 DOI: 10.3390/bioengineering5030056] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 01/19/2023] Open
Abstract
Engineering design of microbioreactors (MBRs) and organ-on-chip (OoC) devices can take advantage of established design science theory, in which systematic evaluation of functional concepts and user requirements are analyzed. This is commonly referred to as a conceptual design. This review article compares how common conceptual design principles are applicable to MBR and OoC devices. The complexity of this design, which is exemplified by MBRs for scaled-down cell cultures in bioprocess development and drug testing in OoCs for heart and eye, is discussed and compared with previous design solutions of MBRs and OoCs, from the perspective of how similarities in understanding design from functionality and user purpose perspectives can more efficiently be exploited. The review can serve as a guideline and help the future design of MBR and OoC devices for cell culture studies.
Collapse
|
19
|
Rehnelt S, Malan D, Juhasz K, Wolters B, Doerr L, Beckler M, Kettenhofen R, Bohlen H, Bruegmann T, Sasse P. Frequency-Dependent Multi-Well Cardiotoxicity Screening Enabled by Optogenetic Stimulation. Int J Mol Sci 2017; 18:E2634. [PMID: 29211031 PMCID: PMC5751237 DOI: 10.3390/ijms18122634] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 11/17/2022] Open
Abstract
Side effects on cardiac ion channels causing lethal arrhythmias are one major reason for drug withdrawals from the market. Field potential (FP) recording from cardiomyocytes, is a well-suited tool to assess such cardiotoxic effects of drug candidates in preclinical drug development, but it is currently limited to the spontaneous beating of the cardiomyocytes and manual analysis. Herein, we present a novel optogenetic cardiotoxicity screening system suited for the parallel automated frequency-dependent analysis of drug effects on FP recorded from human-induced pluripotent stem cell-derived cardiomyocytes. For the expression of the light-sensitive cation channel Channelrhodopsin-2, we optimised protocols using virus transduction or transient mRNA transfection. Optical stimulation was performed with a new light-emitting diode lid for a 96-well FP recording system. This enabled reliable pacing at physiologically relevant heart rates and robust recording of FP. Thereby we detected rate-dependent effects of drugs on Na⁺, Ca2+ and K⁺ channel function indicated by FP prolongation, FP shortening and the slowing of the FP downstroke component, as well as generation of afterdepolarisations. Taken together, we present a scalable approach for preclinical frequency-dependent screening of drug effects on cardiac electrophysiology. Importantly, we show that the recording and analysis can be fully automated and the technology is readily available using commercial products.
Collapse
Affiliation(s)
- Susanne Rehnelt
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Daniela Malan
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Krisztina Juhasz
- Nanion Technologies GmbH, 80636 Munich, Germany.
- Present address: Institute for Nanoelectronics, Department of Electrical Engineering and Information Technology, Technische Universität München, 80339 Munich, Germany.
| | - Benjamin Wolters
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Leo Doerr
- Nanion Technologies GmbH, 80636 Munich, Germany.
| | | | - Ralf Kettenhofen
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Heribert Bohlen
- Part of the Ncardia Group, Axiogenesis AG, 50829 Cologne, Germany.
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
- Research Training Group 1873, University of Bonn, 53127 Bonn, Germany.
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| |
Collapse
|
20
|
Christoffersson J, van Noort D, Mandenius CF. Developing organ-on-a-chip concepts using bio-mechatronic design methodology. Biofabrication 2017; 9:025023. [PMID: 28485301 DOI: 10.1088/1758-5090/aa71ca] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mechatronic design is an engineering methodology for conceiving, configuring and optimising the design of a technical device or product to the needs and requirements of the final user. In this article, we show how the basic principles of this methodology can be exploited for in vitro cell cultures-often referred to as organ-on-a-chip devices. Due to the key role of the biological cells, we have introduced the term bio-mechatronic design, to highlight the complexity of designing a system that should integrate biology, mechanics and electronics in the same device structure. The strength of the mechatronic design is to match the needs of the potential users to a systematic evaluation of overall functional design alternative. It may be especially attractive for organs-on-chips where biological constituents such as cells and tissues in 3D settings and in a fluidic environment should be compared, screened and selected. Through this approach, design solutions ranked to customer needs are generated according to specified criteria, thereby defining the key constraints of the fabrication. As an example, the bio-mechatronic methodology is applied to a liver-on-a-chip based on information extrapolated from previous theoretical and experimental knowledge. It is concluded that the methodology can generate new fabrication solutions for devices, as well as efficient guidelines for refining the design and fabrication of many of today's organ-on-a-chip devices.
Collapse
|
21
|
Tailoring cardiac environment in microphysiological systems: an outlook on current and perspective heart-on-chip platforms. Future Sci OA 2017; 3:FSO191. [PMID: 28670478 PMCID: PMC5481859 DOI: 10.4155/fsoa-2017-0024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 11/17/2022] Open
|
22
|
Pesl M, Pribyl J, Acimovic I, Vilotic A, Jelinkova S, Salykin A, Lacampagne A, Dvorak P, Meli AC, Skladal P, Rotrekl V. Atomic force microscopy combined with human pluripotent stem cell derived cardiomyocytes for biomechanical sensing. Biosens Bioelectron 2016; 85:751-757. [DOI: 10.1016/j.bios.2016.05.073] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/11/2016] [Accepted: 05/23/2016] [Indexed: 11/16/2022]
|
23
|
Takasuna K, Asakura K, Araki S, Ando H, Kazusa K, Kitaguchi T, Kunimatsu T, Suzuki S, Miyamoto N. Comprehensive in vitro cardiac safety assessment using human stem cell technology: Overview of CSAHi HEART initiative. J Pharmacol Toxicol Methods 2016; 83:42-54. [PMID: 27646297 DOI: 10.1016/j.vascn.2016.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 08/23/2016] [Accepted: 09/15/2016] [Indexed: 01/21/2023]
Abstract
Recent increasing evidence suggests that the currently-used platforms in vitro IKr and APD, and/or in vivo QT assays are not fully predictive for TdP, and do not address potential arrhythmia (VT and/or VF) induced by diverse mechanisms of action. In addition, other cardiac safety liabilities such as functional dysfunction of excitation-contraction coupling (contractility) and structural damage (morphological damage to cardiomyocytes) are also major causes of drug attrition, but current in vitro assays do not cover all these liabilities. We organized the Consortium for Safety Assessment using Human iPS cells (CSAHi; http://csahi.org/en/), based on the Japan Pharmaceutical Manufacturers Association (JPMA), to verify the application of human iPS/ES cell-derived cardiomyocytes in drug safety evaluation. The main goal of the CSAHi HEART team has been to propose comprehensive screening strategies to predict a diverse range of cardiotoxicities by using recently introduced platforms (multi-electrode array (MEA), patch clamp, cellular impedance, motion field imaging [MFI], and Ca transient systems) while identifying the strengths and weaknesses of each. Our study shows that hiPS-CMs used in these platforms have pharmacological responses more relevant to humans in comparison with existent hERG, APD or Langendorff (MAPD/contraction) assays, and not only MEA but also other methods such as impedance, MFI, and Ca transient systems would offer paradigm changes of platforms for predicting drug-induced QT risk and/or arrhythmia or contractile dysfunctions. Furthermore, we propose a potential multi-parametric platform in which field potential (MEA)-Ca transient-contraction (MFI) could be evaluated simultaneously as an ideal novel platform for predicting a diversity of cardiac toxicities, namely whole effects on the excitation-contraction cascade.
Collapse
Affiliation(s)
- Kiyoshi Takasuna
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan; Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan.
| | - Keiichi Asakura
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Discovery Research Labs., Nippon Shinyaku Co., Ltd., Kyoto, Japan
| | - Seiichi Araki
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Safety Research Department, ASKA Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Hiroyuki Ando
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Safety Research Laboratories, Ono Pharmaceutical Co., Ltd., Fukui, Japan
| | - Katsuyuki Kazusa
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Drug Safety Research Laboratories, Astellas Pharma Inc., Osaka, Japan
| | - Takashi Kitaguchi
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Discovery Research, Mochida Pharmaceutical Co., Ltd., Shizuoka, Japan
| | - Takeshi Kunimatsu
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Preclinical Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Shinobu Suzuki
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Pharmacokinetics and Non-Clinical Safety Dept., Nippon Boehringer Ingelheim Co., Ltd., Hyogo, Japan
| | - Norimasa Miyamoto
- Japan Pharmaceutical Manufacturers Association Drug Evaluation Committee, Non-Clinical Evaluation Expert Committee, TF2, Japan; Consortium for Safety Assessment using Human iPS Cells (CSAHi): HEART team, Japan; Biopharmaceutical Assessments Core Function Unit Medicine Development Center Eisai Co., Ltd., Eisai Co., Ltd., Ibaraki, Japan
| |
Collapse
|
24
|
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: 587] [Impact Index Per Article: 65.2] [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
|
25
|
Enright HA, Felix SH, Fischer NO, Mukerjee EV, Soscia D, Mcnerney M, Kulp K, Zhang J, Page G, Miller P, Ghetti A, Wheeler EK, Pannu S. Long-term non-invasive interrogation of human dorsal root ganglion neuronal cultures on an integrated microfluidic multielectrode array platform. Analyst 2016; 141:5346-57. [PMID: 27351032 DOI: 10.1039/c5an01728a] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Scientific studies in drug development and toxicology rely heavily on animal models, which often inaccurately predict the true response for human exposure. This may lead to unanticipated adverse effects or misidentified risks that result in, for example, drug candidate elimination. The utilization of human cells and tissues for in vitro physiological platforms has become a growing area of interest to bridge this gap and to more accurately predict human responses to drugs and toxins. The effects of new drugs and toxins on the peripheral nervous system are often investigated with neurons isolated from dorsal root ganglia (DRG), typically with one-time measurement techniques such as patch clamping. Here, we report the use of our multi-electrode array (MEA) platform for long-term noninvasive assessment of human DRG cell health and function. In this study, we acquired simultaneous optical and electrophysiological measurements from primary human DRG neurons upon chemical stimulation repeatedly through day in vitro (DIV) 23. Distinct chemical signatures were noted for the cellular responses evoked by each chemical stimulus. Additionally, the cell viability and function of the human DRG neurons were consistent through DIV 23. To the best of our knowledge, this is the first report on long-term measurements of the cell health and function of human DRG neurons on a MEA platform. Future generations will include higher electrode numbers in customized arrangements as well as integration with different tissue types on a single device. This platform will provide a valuable testing tool for both rodent and human cells, enabling a more comprehensive risk assessment for drug candidates and toxicants.
Collapse
Affiliation(s)
- H A Enright
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
CSAHi study: Evaluation of multi-electrode array in combination with human iPS cell-derived cardiomyocytes to predict drug-induced QT prolongation and arrhythmia — Effects of 7 reference compounds at 10 facilities. J Pharmacol Toxicol Methods 2016; 78:93-102. [DOI: 10.1016/j.vascn.2015.12.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/29/2015] [Accepted: 12/02/2015] [Indexed: 12/23/2022]
|
27
|
A Microfluidic Bioreactor for Toxicity Testing of Stem Cell Derived 3D Cardiac Bodies. Methods Mol Biol 2016; 1502:159-68. [PMID: 27052611 DOI: 10.1007/7651_2016_340] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Modeling tissues and organs using conventional 2D cell cultures is problematic as the cells rapidly lose their in vivo phenotype. In microfluidic bioreactors the cells reside in microstructures that are continuously perfused with cell culture medium to provide a dynamic environment mimicking the cells natural habitat. These micro scale bioreactors are sometimes referred to as organs-on-chips and are developed in order to improve and extend cell culture experiments. Here, we describe the two manufacturing techniques photolithography and soft lithography that are used in order to easily produce microfluidic bioreactors. The use of these bioreactors is exemplified by a toxicity assessment on 3D clustered human pluripotent stem cells (hPSC)-derived cardiomyocytes by beating frequency imaging.
Collapse
|
28
|
Clements M, Millar V, Williams AS, Kalinka S. Bridging Functional and Structural Cardiotoxicity Assays Using Human Embryonic Stem Cell-Derived Cardiomyocytes for a More Comprehensive Risk Assessment. Toxicol Sci 2015; 148:241-60. [DOI: 10.1093/toxsci/kfv180] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
29
|
Bergström G, Christoffersson J, Schwanke K, Zweigerdt R, Mandenius CF. Stem cell derived in vivo-like human cardiac bodies in a microfluidic device for toxicity testing by beating frequency imaging. LAB ON A CHIP 2015; 15:3242-9. [PMID: 26135270 DOI: 10.1039/c5lc00449g] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Beating in vivo-like human cardiac bodies (CBs) were used in a microfluidic device for testing cardiotoxicity. The CBs, cardiomyocyte cell clusters derived from induced pluripotent stem cells, exhibited typical structural and functional properties of the native human myocardium. The CBs were captured in niches along a perfusion channel in the device. Video imaging was utilized for automatic monitoring of the beating frequency of each individual CB. The device allowed assessment of cardiotoxic effects of drug substances doxorubicin, verapamil and quinidine on the 3D clustered cardiomyocytes. Beating frequency data recorded over a period of 6 hours are presented and compared to literature data. The results indicate that this microfluidic setup with imaging of CB characteristics provides a new opportunity for label-free, non-invasive investigation of toxic effects in a 3D microenvironment.
Collapse
Affiliation(s)
- Gunnar Bergström
- Division of Biotechnology, Dept. of Physics, Chemistry and Biology (IFM), Linköping University, 58183 Linköping, Sweden.
| | | | | | | | | |
Collapse
|
30
|
Huyer LD, Montgomery M, Zhao Y, Xiao Y, Conant G, Korolj A, Radisic M. Biomaterial based cardiac tissue engineering and its applications. Biomed Mater 2015; 10:034004. [PMID: 25989939 PMCID: PMC4464787 DOI: 10.1088/1748-6041/10/3/034004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide, necessitating the development of effective treatment strategies. A myocardial infarction involves the blockage of a coronary artery leading to depletion of nutrient and oxygen supply to cardiomyocytes and massive cell death in a region of the myocardium. Cardiac tissue engineering is the growth of functional cardiac tissue in vitro on biomaterial scaffolds for regenerative medicine application. This strategy relies on the optimization of the complex relationship between cell networks and biomaterial properties. In this review, we discuss important biomaterial properties for cardiac tissue engineering applications, such as elasticity, degradation, and induced host response, and their relationship to engineered cardiac cell environments. With these properties in mind, we also emphasize in vitro use of cardiac tissues for high-throughput drug screening and disease modelling.
Collapse
Affiliation(s)
- Locke Davenport Huyer
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Miles Montgomery
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yimu Zhao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yun Xiao
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Genevieve Conant
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Anastasia Korolj
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network and IBBME, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Kunze A, Steel D, Dahlenborg K, Sartipy P, Svedhem S. Non-Invasive Acoustical sensing of Drug-Induced Effects on the Contractile Machinery of Human Cardiomyocyte Clusters. PLoS One 2015; 10:e0125540. [PMID: 25961711 PMCID: PMC4427273 DOI: 10.1371/journal.pone.0125540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 03/25/2015] [Indexed: 11/19/2022] Open
Abstract
There is an urgent need for improved models for cardiotoxicity testing. Here we propose acoustic sensing applied to beating human cardiomyocyte clusters for non-invasive, surrogate measuring of the QT interval and other characteristics of the contractile machinery. In experiments with the acoustic method quartz crystal microbalance with dissipation monitoring (QCM-D), the shape of the recorded signals was very similar to the extracellular field potential detected in electrochemical experiments, and the expected changes of the QT interval in response to addition of conventional drugs (E-4031 or nifedipine) were observed. Additionally, changes in the dissipation signal upon addition of cytochalasin D were in good agreement with the known, corresponding shortening of the contraction-relaxation time. These findings suggest that QCM-D has great potential as a tool for cardiotoxicological screening, where effects of compounds on the cardiomyocyte contractile machinery can be detected independently of whether the extracellular field potential is altered or not.
Collapse
Affiliation(s)
- Angelika Kunze
- Department of Applied Physics, Chalmers University of Technology, Göteborg, Sweden
| | | | | | - Peter Sartipy
- Cellectis AB, Göteborg, Sweden
- Systems Biology Research Center, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Sofia Svedhem
- Department of Applied Physics, Chalmers University of Technology, Göteborg, Sweden
- * E-mail:
| |
Collapse
|