1
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
2
|
Dong M, Oyunbaatar NE, Kanade PP, Kim DS, Lee DW. Real-Time Monitoring of Changes in Cardiac Contractility Using Silicon Cantilever Arrays Integrated with Strain Sensors. ACS Sens 2021; 6:3556-3563. [PMID: 34554741 DOI: 10.1021/acssensors.1c00486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This paper proposes the use of sensor-integrated silicon cantilever arrays to measure drug-induced cardiac toxicity in real time. The proposed cantilever sensors, unlike the conventional electrophysiological methods, aim to evaluate cardiac toxicity by measuring the contraction force of the cardiomyocytes corresponding to the target drugs. The surface of the silicon cantilever consists of microgrooves to maximize the alignment and the contraction force of the cardiomyocytes. This type of surface pattern also helps in the maturation of the cardiomyocytes by increasing the sarcomere length. The preliminary characterization of the cantilever sensors was performed on the cantilever surface, with the cardiomyocytes seeded with a density of 1000 cells/mm2, and the cardiac contractility was measured as a function of the culture days. The change in the contraction force of the cardiomyocytes due to the drug concentration was successfully measured through the integrated strain sensor in the culture media. The reliability of the sensor-integrated cantilevers and the feasibility of their mass production ensure that they meet the practical requirements in the medical applications.
Collapse
Affiliation(s)
- Mingming Dong
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Nomin-Erdene Oyunbaatar
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Pooja P. Kanade
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Su Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Weon Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju 61186, Korea
- Center for Next Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
3
|
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: 10] [Impact Index Per Article: 2.0] [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
|
4
|
Huang D, Gibeley SB, Xu C, Xiao Y, Celik O, Ginsberg HN, Leong KW. Engineering liver microtissues for disease modeling and regenerative medicine. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909553. [PMID: 33390875 PMCID: PMC7774671 DOI: 10.1002/adfm.201909553] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Indexed: 05/08/2023]
Abstract
The burden of liver diseases is increasing worldwide, accounting for two million deaths annually. In the past decade, tremendous progress has been made in the basic and translational research of liver tissue engineering. Liver microtissues are small, three-dimensional hepatocyte cultures that recapitulate liver physiology and have been used in biomedical research and regenerative medicine. This review summarizes recent advances, challenges, and future directions in liver microtissue research. Cellular engineering approaches are used to sustain primary hepatocytes or produce hepatocytes derived from pluripotent stem cells and other adult tissues. Three-dimensional microtissues are generated by scaffold-free assembly or scaffold-assisted methods such as macroencapsulation, droplet microfluidics, and bioprinting. Optimization of the hepatic microenvironment entails incorporating the appropriate cell composition for enhanced cell-cell interactions and niche-specific signals, and creating scaffolds with desired chemical, mechanical and physical properties. Perfusion-based culture systems such as bioreactors and microfluidic systems are used to achieve efficient exchange of nutrients and soluble factors. Taken together, systematic optimization of liver microtissues is a multidisciplinary effort focused on creating liver cultures and on-chip models with greater structural complexity and physiological relevance for use in liver disease research, therapeutic development, and regenerative medicine.
Collapse
Affiliation(s)
- Dantong Huang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Sarah B. Gibeley
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Ozgenur Celik
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
5
|
Wang Z, Xia F, Labib M, Ahmadi M, Chen H, Das J, Ahmed SU, Angers S, Sargent EH, Kelley SO. Nanostructured Architectures Promote the Mesenchymal-Epithelial Transition for Invasive Cells. ACS NANO 2020; 14:5324-5336. [PMID: 32369335 DOI: 10.1021/acsnano.9b07350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Dynamic modulation of cellular phenotypes between the epithelial and mesenchymal states-the epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET)-plays an important role in cancer progression. Nanoscale topography of culture substrates is known to affect the migration and EMT of cancer cells. However, existing platforms heavily rely on simple geometries such as grooved lines or cylindrical post arrays, which may oversimplify the complex interaction between cells and nanotopography in vivo. Here, we use electrodeposition to construct finely controlled surfaces with biomimetic fractal nanostructures as a means of examining the roles of nanotopography during the EMT/MET process. We found that nanostructures in the size range of 100 to 500 nm significantly promote MET for invasive breast and prostate cancer cells. The "METed" cells acquired distinct expression of epithelial and mesenchymal markers, displayed perturbed morphologies, and exhibited diminished migration and invasion, even after the removal of a nanotopographical stimulus. The phosphorylation of GSK-3 was decreased, which further tuned the expression of Snail and modulated the EMT/MET process. Our findings suggest that invasive cancer cells respond to the geometries and dimensions of complex nanostructured architectures.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
| | - Fan Xia
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Moloud Ahmadi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Haijie Chen
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
| | - Jagotamoy Das
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Sharif U Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Stéphane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, M5S 3G4, Canada
| | - Shana O Kelley
- Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, M5S 3M2, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, M5S 1A8, Canada
| |
Collapse
|
6
|
Obenaus AM, Mollica MY, Sniadecki NJ. (De)form and Function: Measuring Cellular Forces with Deformable Materials and Deformable Structures. Adv Healthc Mater 2020; 9:e1901454. [PMID: 31951099 PMCID: PMC7274881 DOI: 10.1002/adhm.201901454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/04/2019] [Indexed: 12/29/2022]
Abstract
The ability for biological cells to produce mechanical forces is important for the development, function, and homeostasis of tissue. The measurement of cellular forces is not a straightforward task because individual cells are microscopic in size and the forces they produce are at the nanonewton scale. Consequently, studies in cell mechanics rely on advanced biomaterials or flexible structures that permit one to infer these forces by the deformation they impart on the material or structure. Herein, the scientific progression on the use of deformable materials and deformable structures to measure cellular forces are reviewed. The findings and insights made possible with these approaches in the field of cell mechanics are summarized.
Collapse
Affiliation(s)
- Ava M Obenaus
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Molly Y Mollica
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
7
|
Sonmez UM, Coyle S, Taylor RE, LeDuc PR. Polycarbonate Heat Molding for Soft Lithography. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2000241. [PMID: 32227442 DOI: 10.1002/smll.202000241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 06/10/2023]
Abstract
Soft lithography enables rapid microfabrication of many types of microsystems by replica molding elastomers into master molds. However, master molds can be very costly, hard to fabricate, vulnerable to damage, and have limited casting life. Here, an approach for the multiplication of master molds into monolithic thermoplastic sheets for further soft lithographic fabrication is introduced. The technique is tested with master molds fabricated through photolithography, mechanical micromilling as well as 3D printing, and the results are demonstrated. Microstructures with submicron feature sizes and high aspect ratios are successfully copied. The copying fidelity of the technique is quantitatively characterized and the microfluidic devices fabricated through this technique are functionally tested. This approach is also used to combine different master molds with up to 19 unique geometries into a single monolithic copy mold in a single step displaying the effectiveness of the copying technique over a large footprint area to scale up the microfabrication. This microfabrication technique can be performed outside the cleanroom without using any sophisticated equipment, suggesting a simple way for high-throughput rigid monolithic mold fabrication that can be used in analytical chemistry studies, biomedical research, and microelectromechanical systems.
Collapse
Affiliation(s)
- Utku M Sonmez
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Stephen Coyle
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Rebecca E Taylor
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Philip R LeDuc
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| |
Collapse
|
8
|
Blair CA, Pruitt BL. Mechanobiology Assays with Applications in Cardiomyocyte Biology and Cardiotoxicity. Adv Healthc Mater 2020; 9:e1901656. [PMID: 32270928 PMCID: PMC7480481 DOI: 10.1002/adhm.201901656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are the motor units that drive the contraction and relaxation of the heart. Traditionally, testing of drugs for cardiotoxic effects has relied on primary cardiomyocytes from animal models and focused on short-term, electrophysiological, and arrhythmogenic effects. However, primary cardiomyocytes present challenges arising from their limited viability in culture, and tissue from animal models suffers from a mismatch in their physiology to that of human heart muscle. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can address these challenges. They also offer the potential to study not only electrophysiological effects but also changes in cardiomyocyte contractile and mechanical function in response to cardiotoxic drugs. With growing recognition of the long-term cardiotoxic effects of some drugs on subcellular structure and function, there is increasing interest in using hiPSC-CMs for in vitro cardiotoxicity studies. This review provides a brief overview of techniques that can be used to quantify changes in the active force that cardiomyocytes generate and variations in their inherent stiffness in response to cardiotoxic drugs. It concludes by discussing the application of these tools in understanding how cardiotoxic drugs directly impact the mechanobiology of cardiomyocytes and how cardiomyocytes sense and respond to mechanical load at the cellular level.
Collapse
Affiliation(s)
- Cheavar A. Blair
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Beth L. Pruitt
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
9
|
Quirós-Solano WF, Gaio N, Silvestri C, Pandraud G, Dekker R, Sarro PM. Metal and Polymeric Strain Gauges for Si-Based, Monolithically Fabricated Organs-on-Chips. MICROMACHINES 2019; 10:mi10080536. [PMID: 31443200 PMCID: PMC6724067 DOI: 10.3390/mi10080536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 01/10/2023]
Abstract
Organ-on-chip (OOC) is becoming the alternative tool to conventional in vitro screening. Heart-on-chip devices including microstructures for mechanical and electrical stimulation have been demonstrated to be advantageous to study structural organization and maturation of heart cells. This paper presents the development of metal and polymeric strain gauges for in situ monitoring of mechanical strain in the Cytostretch platform for heart-on-chip application. Specifically, the optimization of the fabrication process of metal titanium (Ti) strain gauges and the investigation on an alternative material to improve the robustness and performance of the devices are presented. The transduction behavior and functionality of the devices are successfully proven using a custom-made set-up. The devices showed resistance changes for the pressure range (0-3 kPa) used to stretch the membranes on which heart cells can be cultured. Relative resistance changes of approximately 0.008% and 1.2% for titanium and polymeric strain gauges are respectively reported for membrane deformations up to 5%. The results demonstrate that both conventional IC metals and polymeric materials can be implemented for sensing mechanical strain using robust microfabricated organ-on-chip devices.
Collapse
Affiliation(s)
- William F Quirós-Solano
- Department of Microelectronics, Electronic Components, Technology and Materials (ECTM), Delft University of Technology, Mekelweg 4, 2628 CD Delft, The Netherlands.
- School of Electronics Engineering, Instituto Tecnológico de Costa Rica, A.P. 159, 7050 Cartago, Costa Rica.
- BIOND Solutions B.V., Mekelweg 4, 2628 CD Delft, The Netherlands.
| | - Nikolas Gaio
- Department of Microelectronics, Electronic Components, Technology and Materials (ECTM), Delft University of Technology, Mekelweg 4, 2628 CD Delft, The Netherlands
- BIOND Solutions B.V., Mekelweg 4, 2628 CD Delft, The Netherlands
| | - Cinzia Silvestri
- BIOND Solutions B.V., Mekelweg 4, 2628 CD Delft, The Netherlands
| | - Gregory Pandraud
- Electrical Sustainable Energy, Photovoltaic Materials and Devices (PVMD), Delft University of Technology, Mekelweg 4, 2628 CD Delft, The Netherlands
| | - Ronald Dekker
- Department of Microelectronics, Electronic Components, Technology and Materials (ECTM), Delft University of Technology, Mekelweg 4, 2628 CD Delft, The Netherlands
- Philips Research, High Tech Campus, 5656 AE Eindhoven, The Netherlands
| | - Pasqualina M Sarro
- Department of Microelectronics, Electronic Components, Technology and Materials (ECTM), Delft University of Technology, Mekelweg 4, 2628 CD Delft, The Netherlands
| |
Collapse
|
10
|
Wragg NM, Burke L, Wilson SL. A critical review of current progress in 3D kidney biomanufacturing: advances, challenges, and recommendations. RENAL REPLACEMENT THERAPY 2019. [DOI: 10.1186/s41100-019-0218-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
11
|
Aljohani W, Ullah MW, Zhang X, Yang G. Bioprinting and its applications in tissue engineering and regenerative medicine. Int J Biol Macromol 2018; 107:261-275. [DOI: 10.1016/j.ijbiomac.2017.08.171] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 01/16/2023]
|
12
|
Grespan E, Martewicz S, Serena E, Le Houerou V, Rühe J, Elvassore N. Analysis of Calcium Transients and Uniaxial Contraction Force in Single Human Embryonic Stem Cell-Derived Cardiomyocytes on Microstructured Elastic Substrate with Spatially Controlled Surface Chemistries. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:12190-12201. [PMID: 27643958 DOI: 10.1021/acs.langmuir.6b03138] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The mechanical activity of cardiomyocytes is the result of a process called excitation-contraction coupling (ECC). A membrane depolarization wave induces a transient cytosolic calcium concentration increase that triggers activation of calcium-sensitive contractile proteins, leading to cell contraction and force generation. An experimental setup capable of acquiring simultaneously all ECC features would have an enormous impact on cardiac drug development and disease study. In this work, we develop a microengineered elastomeric substrate with tailor-made surface chemistry to measure simultaneously the uniaxial contraction force and the calcium transients generated by single human cardiomyocytes in vitro. Microreplication followed by photocuring is used to generate an array consisting of elastomeric micropillars. A second photochemical process is employed to spatially control the surface chemistry of the elastomeric pillar. As result, human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can be confined in rectangular cell-adhesive areas, which induce cell elongation and promote suspended cell anchoring between two adjacent micropillars. In this end-to-end conformation, confocal fluorescence microscopy allows simultaneous detection of calcium transients and micropillar deflection induced by a single-cell uniaxial contraction force. Computational finite elements modeling (FEM) and 3D reconstruction of the cell-pillar interface allow force quantification. The platform is used to follow calcium dynamics and contraction force evolution in hESC-CMs cultures over the course of several weeks. Our results show how a biomaterial-based platform can be a versatile tool for in vitro assaying of cardiac functional properties of single-cell human cardiomyocytes, with applications in both in vitro developmental studies and drug screening on cardiac cultures.
Collapse
Affiliation(s)
- Eleonora Grespan
- CNR Institute of Neuroscience , Corso Stati Uniti 4, 35127 Padova, Italy
| | - Sebastian Martewicz
- Department of Industrial Engineering, University of Padova , Via Marzolo 9, 35131 Padova, Italy
- Venetian Institute of Molecular Medicine , Via Orus 2, 35129 Padua, Italy
| | - Elena Serena
- Department of Industrial Engineering, University of Padova , Via Marzolo 9, 35131 Padova, Italy
- Venetian Institute of Molecular Medicine , Via Orus 2, 35129 Padua, Italy
| | - Vincent Le Houerou
- Institute Charles Sadron, University of Strasbourg , 23 rue du Loess, 84047 Strasbourg, France
| | - Jürgen Rühe
- Department for Microsystems Engineering, University of Freiburg , Georges-Köhler Allee 103, 79110 Freiburg, Germany
| | - Nicola Elvassore
- Department of Industrial Engineering, University of Padova , Via Marzolo 9, 35131 Padova, Italy
- Venetian Institute of Molecular Medicine , Via Orus 2, 35129 Padua, Italy
| |
Collapse
|
13
|
Mandrycky C, Wang Z, Kim K, Kim DH. 3D bioprinting for engineering complex tissues. Biotechnol Adv 2016; 34:422-434. [PMID: 26724184 PMCID: PMC4879088 DOI: 10.1016/j.biotechadv.2015.12.011] [Citation(s) in RCA: 912] [Impact Index Per Article: 101.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/10/2015] [Accepted: 12/22/2015] [Indexed: 02/07/2023]
Abstract
Bioprinting is a 3D fabrication technology used to precisely dispense cell-laden biomaterials for the construction of complex 3D functional living tissues or artificial organs. While still in its early stages, bioprinting strategies have demonstrated their potential use in regenerative medicine to generate a variety of transplantable tissues, including skin, cartilage, and bone. However, current bioprinting approaches still have technical challenges in terms of high-resolution cell deposition, controlled cell distributions, vascularization, and innervation within complex 3D tissues. While no one-size-fits-all approach to bioprinting has emerged, it remains an on-demand, versatile fabrication technique that may address the growing organ shortage as well as provide a high-throughput method for cell patterning at the micrometer scale for broad biomedical engineering applications. In this review, we introduce the basic principles, materials, integration strategies and applications of bioprinting. We also discuss the recent developments, current challenges and future prospects of 3D bioprinting for engineering complex tissues. Combined with recent advances in human pluripotent stem cell technologies, 3D-bioprinted tissue models could serve as an enabling platform for high-throughput predictive drug screening and more effective regenerative therapies.
Collapse
Affiliation(s)
- Christian Mandrycky
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Zongjie Wang
- School of Engineering, The University of British Columbia, Kelowna, BC V1V 1V7, Canada
| | - Keekyoung Kim
- School of Engineering, The University of British Columbia, Kelowna, BC V1V 1V7, Canada.
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
14
|
Ribeiro AJS, Denisin AK, Wilson RE, Pruitt BL. For whom the cells pull: Hydrogel and micropost devices for measuring traction forces. Methods 2016; 94:51-64. [PMID: 26265073 PMCID: PMC4746112 DOI: 10.1016/j.ymeth.2015.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 07/10/2015] [Accepted: 08/06/2015] [Indexed: 01/16/2023] Open
Abstract
While performing several functions, adherent cells deform their surrounding substrate via stable adhesions that connect the intracellular cytoskeleton to the extracellular matrix. The traction forces that deform the substrate are studied in mechanotrasduction because they are affected by the mechanics of the extracellular milieu. We review the development and application of two methods widely used to measure traction forces generated by cells on 2D substrates: (i) traction force microscopy with polyacrylamide hydrogels and (ii) calculation of traction forces with arrays of deformable microposts. Measuring forces with these methods relies on measuring substrate displacements and converting them into forces. We describe approaches to determine force from displacements and elaborate on the necessary experimental conditions for this type of analysis. We emphasize device fabrication, mechanical calibration of substrates and covalent attachment of extracellular matrix proteins to substrates as key features in the design of experiments to measure cell traction forces with polyacrylamide hydrogels or microposts. We also report the challenges and achievements in integrating these methods with platforms for the mechanical stimulation of adherent cells. The approaches described here will enable new studies to understand cell mechanical outputs as a function of mechanical inputs and advance the understanding of mechanotransduction mechanisms.
Collapse
Affiliation(s)
- Alexandre J S Ribeiro
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States
| | - Aleksandra K Denisin
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Bioengineering, Stanford University, Stanford, CA 94305, United States
| | - Robin E Wilson
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States
| | - Beth L Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, United States; Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, United States; Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, United States.
| |
Collapse
|
15
|
Beussman KM, Rodriguez ML, Leonard A, Taparia N, Thompson CR, Sniadecki NJ. Micropost arrays for measuring stem cell-derived cardiomyocyte contractility. Methods 2016; 94:43-50. [PMID: 26344757 PMCID: PMC4761463 DOI: 10.1016/j.ymeth.2015.09.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/14/2022] Open
Abstract
Stem cell-derived cardiomyocytes have the potential to be used to study heart disease and maturation, screen drug treatments, and restore heart function. Here, we discuss the procedures involved in using micropost arrays to measure the contractile forces generated by stem cell-derived cardiomyocytes. Cardiomyocyte contractility is needed for the heart to pump blood, so measuring the contractile forces of cardiomyocytes is a straightforward way to assess their function. Microfabrication and soft lithography techniques are utilized to create identical arrays of flexible, silicone microposts from a common master. Micropost arrays are functionalized with extracellular matrix protein to allow cardiomyocytes to adhere to the tips of the microposts. Live imaging is used to capture videos of the deflection of microposts caused by the contraction of the cardiomyocytes. Image analysis code provides an accurate means to quantify these deflections. The contractile forces produced by a beating cardiomyocyte are calculated by modeling the microposts as cantilever beams. We have used this assay to assess techniques for improving the maturation and contractile function of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Kevin M Beussman
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Marita L Rodriguez
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Nikita Taparia
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Curtis R Thompson
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Hartman ME, Dai DF, Laflamme MA. Human pluripotent stem cells: Prospects and challenges as a source of cardiomyocytes for in vitro modeling and cell-based cardiac repair. Adv Drug Deliv Rev 2016; 96:3-17. [PMID: 25980938 DOI: 10.1016/j.addr.2015.05.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/27/2015] [Accepted: 05/07/2015] [Indexed: 12/13/2022]
Abstract
Human pluripotent stem cells (PSCs) represent an attractive source of cardiomyocytes with potential applications including disease modeling, drug discovery and safety screening, and novel cell-based cardiac therapies. Insights from embryology have contributed to the development of efficient, reliable methods capable of generating large quantities of human PSC-cardiomyocytes with cardiac purities ranging up to 90%. However, for human PSCs to meet their full potential, the field must identify methods to generate cardiomyocyte populations that are uniform in subtype (e.g. homogeneous ventricular cardiomyocytes) and have more mature structural and functional properties. For in vivo applications, cardiomyocyte production must be highly scalable and clinical grade, and we will need to overcome challenges including graft cell death, immune rejection, arrhythmogenesis, and tumorigenic potential. Here we discuss the types of human PSCs, commonly used methods to guide their differentiation into cardiomyocytes, the phenotype of the resultant cardiomyocytes, and the remaining obstacles to their successful translation.
Collapse
|
17
|
Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proc Natl Acad Sci U S A 2015; 112:12705-10. [PMID: 26417073 DOI: 10.1073/pnas.1508073112] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Single cardiomyocytes contain myofibrils that harbor the sarcomere-based contractile machinery of the myocardium. Cardiomyocytes differentiated from human pluripotent stem cells (hPSC-CMs) have potential as an in vitro model of heart activity. However, their fetal-like misalignment of myofibrils limits their usefulness for modeling contractile activity. We analyzed the effects of cell shape and substrate stiffness on the shortening and movement of labeled sarcomeres and the translation of sarcomere activity to mechanical output (contractility) in live engineered hPSC-CMs. Single hPSC-CMs were cultured on polyacrylamide substrates of physiological stiffness (10 kPa), and Matrigel micropatterns were used to generate physiological shapes (2,000-µm(2) rectangles with length:width aspect ratios of 5:1-7:1) and a mature alignment of myofibrils. Translation of sarcomere shortening to mechanical output was highest in 7:1 hPSC-CMs. Increased substrate stiffness and applied overstretch induced myofibril defects in 7:1 hPSC-CMs and decreased mechanical output. Inhibitors of nonmuscle myosin activity repressed the assembly of myofibrils, showing that subcellular tension drives the improved contractile activity in these engineered hPSC-CMs. Other factors associated with improved contractility were axially directed calcium flow, systematic mitochondrial distribution, more mature electrophysiology, and evidence of transverse-tubule formation. These findings support the potential of these engineered hPSC-CMs as powerful models for studying myocardial contractility at the cellular level.
Collapse
|
18
|
Microsphere-assisted fabrication of high aspect-ratio elastomeric micropillars and waveguides. Nat Commun 2015; 5:3324. [PMID: 24525627 DOI: 10.1038/ncomms4324] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 01/27/2014] [Indexed: 11/08/2022] Open
Abstract
High aspect-ratio micropillars are in strong demand for microtechnology, but their realization remains a difficult challenge, especially when attempted with soft materials. Here we present a direct drawing-based technique for fabricating micropillars with poly(dimethylsiloxane). Despite the material's extreme softness, our technique enables routine realization of micropillars exceeding 2,000 μm in height and 100 in aspect-ratio. It also supports in situ integration of microspheres at the tips of the micropillars. As a validation of the new structure's utility, we configure it into airflow sensors, in which the micropillars and microspheres function as flexible upright waveguides and self-aligned reflectors, respectively. High-level bending of the micropillar under an airflow and its optical read-out enables mm s(-1) scale-sensing resolution. This new scheme, which uniquely integrates high aspect-ratio elastomeric micropillars and microspheres self-aligned to them, could widen the scope of soft material-based microdevice technology.
Collapse
|
19
|
Rodriguez ML, Graham BT, Pabon LM, Han SJ, Murry CE, Sniadecki NJ. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J Biomech Eng 2015; 136:051005. [PMID: 24615475 DOI: 10.1115/1.4027145] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Abstract
Human stem cell-derived cardiomyocytes hold promise for heart repair, disease modeling, drug screening, and for studies of developmental biology. All of these applications can be improved by assessing the contractility of cardiomyocytes at the single cell level. We have developed an in vitro platform for assessing the contractile performance of stem cell-derived cardiomyocytes that is compatible with other common endpoints such as microscopy and molecular biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were seeded onto elastomeric micropost arrays in order to characterize the contractile force, velocity, and power produced by these cells. We assessed contractile function by tracking the deflection of microposts beneath an individual hiPSC-CM with optical microscopy. Immunofluorescent staining of these cells was employed to assess their spread area, nucleation, and sarcomeric structure on the microposts. Following seeding of hiPSC-CMs onto microposts coated with fibronectin, laminin, and collagen IV, we found that hiPSC-CMs on laminin coatings demonstrated higher attachment, spread area, and contractile velocity than those seeded on fibronectin or collagen IV coatings. Under optimized conditions, hiPSC-CMs spread to an area of approximately 420 μm2, generated systolic forces of approximately 15 nN/cell, showed contraction and relaxation rates of 1.74 μm/s and 1.46 μm/s, respectively, and had a peak contraction power of 29 fW. Thus, elastomeric micropost arrays can be used to study the contractile strength and kinetics of hiPSC-CMs. This system should facilitate studies of hiPSC-CM maturation, disease modeling, and drug screens as well as fundamental studies of human cardiac contraction.
Collapse
|
20
|
Abstract
Biological mechano-transduction and force-dependent changes scale from protein conformation (â„« to nm) to cell organization and multi-cell function (mm to cm) to affect cell organization, fate, and homeostasis. External forces play complex roles in cell organization, fate, and homeostasis. Changes in these forces, or how cells respond to them, can result in abnormal embryonic development and diseases in adults. How cells sense and respond to these mechanical stimuli requires an understanding of the biophysical principles that underlie changes in protein conformation and result in alterations in the organization and function of cells and tissues. Here, we discuss mechano-transduction as it applies to protein conformation, cellular organization, and multi-cell (tissue) function.
Collapse
Affiliation(s)
- Beth L. Pruitt
- Department of Mechanical Engineering, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - Alexander R. Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - William I. Weis
- Department of Structural Biology, Stanford University, Stanford, California, United States of America
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| | - W. James Nelson
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Department of Biology, Stanford University, Stanford, California, United States of America
- * E-mail: (BLP); (ARD); (WIW); (WJN)
| |
Collapse
|
21
|
Ribeiro AS, Zaleta-Rivera K, Ashley EA, Pruitt BL. Stable, covalent attachment of laminin to microposts improves the contractility of mouse neonatal cardiomyocytes. ACS APPLIED MATERIALS & INTERFACES 2014; 6:15516-26. [PMID: 25133578 PMCID: PMC4160263 DOI: 10.1021/am5042324] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The mechanical output of contracting cardiomyocytes, the muscle cells of the heart, relates to healthy and disease states of the heart. Culturing cardiomyocytes on arrays of elastomeric microposts can enable inexpensive and high-throughput studies of heart disease at the single-cell level. However, cardiomyocytes weakly adhere to these microposts, which limits the possibility of using biomechanical assays of single cardiomyocytes to study heart disease. We hypothesized that a stable covalent attachment of laminin to the surface of microposts improves cardiomyocyte contractility. We cultured cells on polydimethylsiloxane microposts with laminin covalently bonded with the organosilanes 3-glycidoxypropyltrimethoxysilane and 3-aminopropyltriethoxysilane with glutaraldehyde. We measured displacement of microposts induced by the contractility of mouse neonatal cardiomyocytes, which attach better than mature cardiomyocytes to substrates. We observed time-dependent changes in contractile parameters such as micropost deformation, contractility rates, contraction and relaxation speeds, and the times of contractions. These parameters were affected by the density of laminin on microposts and by the stability of laminin binding to micropost surfaces. Organosilane-mediated binding resulted in higher laminin surface density and laminin binding stability. 3-glycidoxypropyltrimethoxysilane provided the highest laminin density but did not provide stable protein binding with time. Higher surface protein binding stability and strength were observed with 3-aminopropyltriethoxysilane with glutaraldehyde. In cultured cardiomyocytes, contractility rate, contraction speeds, and contraction time increased with higher laminin stability. Given these variations in contractile function, we conclude that binding of laminin to microposts via 3-aminopropyltriethoxysilane with glutaraldehyde improves contractility observed by an increase in beating rate and contraction speed as it occurs during the postnatal maturation of cardiomyocytes. This approach is promising for future studies to mimic in vivo tissue environments.
Collapse
Affiliation(s)
- Alexandre
J. S. Ribeiro
- Department of Mechanical Engineering and Stanford Cardiovascular Institute, Stanford University, Stanford, California United
States
| | - Kathia Zaleta-Rivera
- Department of Mechanical Engineering and Stanford Cardiovascular Institute, Stanford University, Stanford, California United
States
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California United States
| | - Euan A. Ashley
- Department of Mechanical Engineering and Stanford Cardiovascular Institute, Stanford University, Stanford, California United
States
- Department of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California United States
| | - Beth L. Pruitt
- Department of Mechanical Engineering and Stanford Cardiovascular Institute, Stanford University, Stanford, California United
States
- E-mail: . Fax: +1 650 725 1587. Phone: +1 650 723 2300
| |
Collapse
|
22
|
Construction of a microrobot system using magnetotactic bacteria for the separation of Staphylococcus aureus. Biomed Microdevices 2014; 16:761-70. [DOI: 10.1007/s10544-014-9880-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
23
|
Abstract
OPINION STATEMENT Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) represent a powerful new model system to study the basic mechanisms of inherited cardiomyopathies. hiPSC-CMs have been utilized to model several cardiovascular diseases, achieving the most success in the inherited arrhythmias, including long QT and Timothy syndromes (Moretti et al. N Engl J Med. 363:1397-409, 2010; Yazawa et al. Nature. 471:230-4, 2011) and arrhythmogenic right ventricular dysplasia (ARVD) (Ma et al. Eur Heart J. 34:1122-33, 2013). Recently, studies have applied hiPSC-CMs to the study of both dilated (DCM) (Sun et al. Sci Transl Med. 4:130ra47, 2012) and hypertrophic (HCM) cardiomyopathies (Lan et al. Cell Stem Cell. 12:101-13, 2013; Carvajal-Vergara et al. Nature. 465:808-12, 2010), providing new insights into basic mechanisms of disease. However, hiPSC-CMs do not recapitulate many of the structural and functional aspects of mature human cardiomyocytes, instead mirroring an immature - embryonic or fetal - phenotype. Much work remains in order to better understand these differences, as well as to develop methods to induce hiPSC-CMs into a fully mature phenotype. Despite these limitations, hiPSC-CMs represent the best current in vitro correlate of the human heart and an invaluable tool in the search for mechanisms underlying cardiomyopathy and for screening new pharmacologic therapies.
Collapse
|
24
|
Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H, Regnier M, Sniadecki NJ, Ruohola-Baker H, Murry CE. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J Mol Cell Cardiol 2014; 72:296-304. [PMID: 24735830 DOI: 10.1016/j.yjmcc.2014.04.005] [Citation(s) in RCA: 316] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/15/2014] [Accepted: 04/05/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) have great potential as a cell source for therapeutic applications such as regenerative medicine, disease modeling, drug screening, and toxicity testing. This potential is limited, however, by the immature state of the cardiomyocytes acquired using current protocols. Tri-iodo-l-thyronine (T3) is a growth hormone that is essential for optimal heart growth. In this study, we investigated the effect of T3 on hiPSC-CM maturation. METHODS AND RESULTS A one-week treatment with T3 increased cardiomyocyte size, anisotropy, and sarcomere length. T3 treatment was associated with reduced cell cycle activity, manifest as reduced DNA synthesis and increased expression of the cyclin-dependent kinase inhibitor p21. Contractile force analyses were performed on individual cardiomyocytes using arrays of microposts, revealing an almost two-fold higher force per-beat after T3 treatment and also an enhancement in contractile kinetics. This improvement in force generation was accompanied by an increase in rates of calcium release and reuptake, along with a significant increase in sarcoendoplasmic reticulum ATPase expression. Finally, although mitochondrial genomes were not numerically increased, extracellular flux analysis showed a significant increase in maximal mitochondrial respiratory capacity and respiratory reserve capability after T3 treatment. CONCLUSIONS Using a broad spectrum of morphological, molecular, and functional parameters, we conclude that T3 is a driver for hiPSC-CM maturation. T3 treatment may enhance the utility of hiPSC-CMs for therapy, disease modeling, or drug/toxicity screens.
Collapse
Affiliation(s)
- Xiulan Yang
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Marita Rodriguez
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA
| | - Lil Pabon
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Karin A Fischer
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Hans Reinecke
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | | | - Charles E Murry
- Department of Pathology, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|