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
|
Xu F, Jin H, Liu L, Yang Y, Cen J, Wu Y, Chen S, Sun D. Architecture design and advanced manufacturing of heart-on-a-chip: scaffolds, stimulation and sensors. MICROSYSTEMS & NANOENGINEERING 2024; 10:96. [PMID: 39006908 PMCID: PMC11239895 DOI: 10.1038/s41378-024-00692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 07/16/2024]
Abstract
Heart-on-a-chip (HoC) has emerged as a highly efficient, cost-effective device for the development of engineered cardiac tissue, facilitating high-throughput testing in drug development and clinical treatment. HoC is primarily used to create a biomimetic microphysiological environment conducive to fostering the maturation of cardiac tissue and to gather information regarding the real-time condition of cardiac tissue. The development of architectural design and advanced manufacturing for these "3S" components, scaffolds, stimulation, and sensors is essential for improving the maturity of cardiac tissue cultivated on-chip, as well as the precision and accuracy of tissue states. In this review, the typical structures and manufacturing technologies of the "3S" components are summarized. The design and manufacturing suggestions for each component are proposed. Furthermore, key challenges and future perspectives of HoC platforms with integrated "3S" components are discussed. Architecture design concepts of scaffolds, stimulation and sensors in chips.
Collapse
Affiliation(s)
- Feng Xu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Hang Jin
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Lingling Liu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Yuanyuan Yang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Jianzheng Cen
- Guangdong Provincial People’s Hospital, Guangzhou, 510080 China
| | - Yaobin Wu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Songyue Chen
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| | - Daoheng Sun
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102 China
| |
Collapse
|
3
|
Gokhan I, Blum TS, Campbell SG. Engineered heart tissue: Design considerations and the state of the art. BIOPHYSICS REVIEWS 2024; 5:021308. [PMID: 38912258 PMCID: PMC11192576 DOI: 10.1063/5.0202724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Originally developed more than 20 years ago, engineered heart tissue (EHT) has become an important tool in cardiovascular research for applications such as disease modeling and drug screening. Innovations in biomaterials, stem cell biology, and bioengineering, among other fields, have enabled EHT technologies to recapitulate many aspects of cardiac physiology and pathophysiology. While initial EHT designs were inspired by the isolated-trabecula culture system, current designs encompass a variety of formats, each of which have unique strengths and limitations. In this review, we describe the most common EHT formats, and then systematically evaluate each aspect of their design, emphasizing the rational selection of components for each application.
Collapse
Affiliation(s)
| | - Thomas S. Blum
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | | |
Collapse
|
4
|
Gu B, Han K, Cao H, Huang X, Li X, Mao M, Zhu H, Cai H, Li D, He J. Heart-on-a-chip systems with tissue-specific functionalities for physiological, pathological, and pharmacological studies. Mater Today Bio 2024; 24:100914. [PMID: 38179431 PMCID: PMC10765251 DOI: 10.1016/j.mtbio.2023.100914] [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: 10/05/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in heart-on-a-chip systems hold great promise to facilitate cardiac physiological, pathological, and pharmacological studies. This review focuses on the development of heart-on-a-chip systems with tissue-specific functionalities. For one thing, the strategies for developing cardiac microtissues on heart-on-a-chip systems that closely mimic the structures and behaviors of the native heart are analyzed, including the imitation of cardiac structural and functional characteristics. For another, the development of techniques for real-time monitoring of biophysical and biochemical signals from cardiac microtissues on heart-on-a-chip systems is introduced, incorporating cardiac electrophysiological signals, contractile activity, and biomarkers. Furthermore, the applications of heart-on-a-chip systems in intelligent cardiac studies are discussed regarding physiological/pathological research and pharmacological assessment. Finally, the future development of heart-on-a-chip toward a higher level of systematization, integration, and maturation is proposed.
Collapse
Affiliation(s)
- Bingsong Gu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Kang Han
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hanbo Cao
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Xinxin Huang
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Xiao Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Mao Mao
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hui Zhu
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Hu Cai
- Shaanxi Provincial Institute for Food and Drug Control, Xi’ an, 710065, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| | - Jiankang He
- State Key Laboratory for Manufacturing System Engineering, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi’ an, 710049, China
- National Innovation Platform (Center) for Industry-Education Integration of Medical Technology, Xi'an Jiaotong University, China
| |
Collapse
|
5
|
Strohm EM, Callaghan NI, Ding Y, Latifi N, Rafatian N, Funakoshi S, Fernandes I, Reitz CJ, Di Paola M, Gramolini AO, Radisic M, Keller G, Kolios MC, Simmons CA. Noninvasive Quantification of Contractile Dynamics in Cardiac Cells, Spheroids, and Organs-on-a-Chip Using High-Frequency Ultrasound. ACS NANO 2024; 18:314-327. [PMID: 38147684 DOI: 10.1021/acsnano.3c06325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Cell-based models that mimic in vivo heart physiology are poised to make significant advances in cardiac disease modeling and drug discovery. In these systems, cardiomyocyte (CM) contractility is an important functional metric, but current measurement methods are inaccurate and low-throughput or require complex setups. To address this need, we developed a standalone noninvasive, label-free ultrasound technique operating at 40-200 MHz to measure the contractile kinetics of cardiac models, ranging from single adult CMs to 3D microtissue constructs in standard cell culture formats. The high temporal resolution of 1000 fps resolved the beat profile of single mouse CMs paced at up to 9 Hz, revealing limitations of lower speed optical based measurements to resolve beat kinetics or characterize aberrant beats. Coupling of ultrasound with traction force microscopy enabled the measurement of the CM longitudinal modulus and facile estimation of adult mouse CM contractile forces of 2.34 ± 1.40 μN, comparable to more complex measurement techniques. Similarly, the beat rate, rhythm, and drug responses of CM spheroid and microtissue models were measured, including in configurations without optical access. In conclusion, ultrasound can be used for the rapid characterization of CM contractile function in a wide range of commonly studied configurations ranging from single cells to 3D tissue constructs using standard well plates and custom microdevices, with applications in cardiac drug discovery and cardiotoxicity evaluation.
Collapse
Affiliation(s)
- Eric M Strohm
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, M5S 3G8, Canada
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
| | - Neal I Callaghan
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
| | - Yu Ding
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
| | - Neda Latifi
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, M5S 3G8, Canada
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
| | - Naimeh Rafatian
- Toronto General Hospital Research Institute, Toronto, M5G 2C4, Canada
| | - Shunsuke Funakoshi
- McEwen Stem Cell Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Ian Fernandes
- McEwen Stem Cell Institute, University Health Network, Toronto, M5G 1L7, Canada
| | - Cristine J Reitz
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Michelle Di Paola
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
- Toronto General Hospital Research Institute, Toronto, M5G 2C4, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, M5S 3E5, Canada
| | - Gordon Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, M5B 2K3, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, M5S 3G8, Canada
- Translational Biology and Engineering Program, Ted Rogers Center for Heart Research, Toronto, M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S 3G9, Canada
| |
Collapse
|
6
|
Sun H, Kim DS, Shanmugasundaram A, Kim JY, Kim ES, Lee BK, Lee DW. Enhancing cardiomyocytes contraction force measuring in drug testing: Integration of a highly sensitive single-crystal silicon strain sensor into SU-8 cantilevers. Biosens Bioelectron 2024; 243:115756. [PMID: 37898097 DOI: 10.1016/j.bios.2023.115756] [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: 08/02/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/30/2023]
Abstract
The development of efficient tools for predicting drug-induced cardiotoxicity in the preclinical phase would greatly benefit the drug development process. This study presents an SU-8 cantilever integrated with a single-crystal silicon strain sensor to enhance force sensitivity in toxicity screening methods based on changes in the contraction force of cardiomyocytes. The proposed cantilever device enables real-time measurements of cardiomyocytes contraction force with high sensitivity, thereby facilitating the assessment of drug cardiotoxicity. The experimental results obtained herein demonstrate the responsiveness of the proposed platform in detecting forces smaller than 0.02 μN with a force sensitivity that is nearly 17 times higher than those of conventional metal-based strain sensors. Moreover, the integration of strain sensors demonstrates the potential for manufacturing cantilever arrays that can be used in high-throughput screening applications. The developed methodology successfully facilitates in vitro culturing of cardiomyocytes and allows for continuous monitoring of their contraction force. The practical applicability of the proposed platform is further validated through cardiotoxicity analysis. The cultured cardiomyocytes are treated with two cardiovascular drugs, namely verapamil (an L-type calcium channel blocker) and isoproterenol (a sympathomimetic drug targeting β1 and β2 adrenergic receptors), to analyze the drug induced effects in the cardiomyocytes.
Collapse
Affiliation(s)
- Haolan Sun
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Dong-Su Kim
- Green Energy & Nano Technology R&D Group, Korea Institute of Industrial Technology (KITECH), Gwangju, 61012, Republic of Korea
| | - Arunkumar Shanmugasundaram
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jong-Yun Kim
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Eung-Sam Kim
- School of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Bong-Kee Lee
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Dong-Weon Lee
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju, 61186, Republic of Korea; Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
7
|
Rashid SA, Dong Y, Ogasawara H, Vierengel M, Essien ME, Salaita K. All-Covalent Nuclease-Resistant and Hydrogel-Tethered DNA Hairpin Probes Map pN Cell Traction Forces. ACS APPLIED MATERIALS & INTERFACES 2023; 15:33362-33372. [PMID: 37409737 PMCID: PMC10360067 DOI: 10.1021/acsami.3c04826] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Cells sense and respond to the physical properties of their environment through receptor-mediated signaling, a process known as mechanotransduction, which can modulate critical cellular functions such as proliferation, differentiation, and survival. At the molecular level, cell adhesion receptors, such as integrins, transmit piconewton (pN)-scale forces to the extracellular matrix, and the magnitude of the force plays a critical role in cell signaling. The most sensitive approach to measuring integrin forces involves DNA hairpin-based sensors, which are used to quantify and map forces in living cells. Despite the broad use of DNA hairpin sensors to study a variety of mechanotransduction processes, these sensors are typically anchored to rigid glass slides, which are orders of magnitude stiffer than the extracellular matrix and hence modulate native biological responses. Here, we have developed nuclease-resistant DNA hairpin probes that are all covalently tethered to PEG hydrogels to image cell traction forces on physiologically relevant substrate stiffness. Using HeLa cells as a model cell line, we show that the molecular forces transmitted by integrins are highly sensitive to the bulk modulus of the substrate, and cells cultured on the 6 and 13 kPa gels produced a greater number of hairpin unfolding events compared to the 2 kPa substrates. Tension signals are spatially colocalized with pY118-paxillin, confirming focal adhesion-mediated probe opening. Additionally, we found that integrin forces are greater than 5.8 pN but less than 19 pN on 13 kPa gels. This work provides a general strategy to integrate molecular tension probes into hydrogels, which can better mimic in vivo mechanotransduction.
Collapse
Affiliation(s)
- Sk Aysha Rashid
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Hiroaki Ogasawara
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Maia Vierengel
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mark Edoho Essien
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
8
|
Chen H, Luo Z, Lin X, Zhu Y, Zhao Y. Sensors-integrated organ-on-a-chip for biomedical applications. NANO RESEARCH 2023; 16:1-28. [PMID: 37359077 PMCID: PMC10130312 DOI: 10.1007/s12274-023-5651-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 06/28/2023]
Abstract
As a promising new micro-physiological system, organ-on-a-chip has been widely utilized for in vitro pharmaceutical study and tissues engineering based on the three-dimensional constructions of tissues/organs and delicate replication of in vivo-like microenvironment. To better observe the biological processes, a variety of sensors have been integrated to realize in-situ, real-time, and sensitive monitoring of critical signals for organs development and disease modeling. Herein, we discuss the recent research advances made with respect to sensors-integrated organ-on-a-chip in this overall review. Firstly, we briefly explore the underlying fabrication procedures of sensors within microfluidic platforms and several classifications of sensory principles. Then, emphasis is put on the highlighted applications of different types of organ-on-a-chip incorporated with various sensors. Last but not least, perspective on the remaining challenges and future development of sensors-integrated organ-on-a-chip are presented.
Collapse
Affiliation(s)
- Hanxu Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Zhiqiang Luo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yujuan Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096 China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001 China
| |
Collapse
|
9
|
Ikegami R, Tsukagoshi T, Matsudaira K, Shoji KH, Takahashi H, Nguyen TV, Tamamoto T, Noda K, Koyanagi K, Oshima T, Shimoyama I. Temperature Dependence of the Beating Frequency of hiPSC-CMs Using a MEMS Force Sensor. SENSORS (BASEL, SWITZERLAND) 2023; 23:3370. [PMID: 37050430 PMCID: PMC10098744 DOI: 10.3390/s23073370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
It is expected that human iPS cell-derived cardiomyocytes (hiPSC-CMs) can be used to treat serious heart diseases. However, the properties and functions of human adult cardiomyocytes and hiPSC-CMs, including cell maturation, differ. In this study, we focused on the temperature dependence of hiPSC-CMs by integrating the temperature regulation system into our sensor platform, which can directly and quantitatively measure their mechanical motion. We measured the beating frequency of hiPSC-CMs at different environmental temperatures and found that the beating frequency increased as the temperature increased. Although the rate at which the beating frequency increased with temperature varied, the temperature at which the beating stopped was relatively stable at approximately 20 °C. The stopping of beating at this temperature was stable, even in immature hiPSC-CMs, and was considered to be a primitive property of cardiomyocytes.
Collapse
Affiliation(s)
- Ryota Ikegami
- Department of Intelligent Robotics, Faculty of Engineering, Toyama Prefectural University, Imizu 939-0398, Japan
| | - Takuya Tsukagoshi
- Department of Intelligent Robotics, Faculty of Engineering, Toyama Prefectural University, Imizu 939-0398, Japan
| | - Kenei Matsudaira
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Kayoko Hirayama Shoji
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Hidetoshi Takahashi
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Thanh-Vinh Nguyen
- Sensing System Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8564, Japan
| | - Takumi Tamamoto
- Department of Intelligent Mechanical Engineering, Faculty of Engineering, Fukuoka Institute of Technology, Fukuoka 811-0295, Japan
| | - Kentaro Noda
- Department of Intelligent Robotics, Faculty of Engineering, Toyama Prefectural University, Imizu 939-0398, Japan
| | - Ken’ichi Koyanagi
- Department of Intelligent Robotics, Faculty of Engineering, Toyama Prefectural University, Imizu 939-0398, Japan
| | - Toru Oshima
- Department of Intelligent Robotics, Faculty of Engineering, Toyama Prefectural University, Imizu 939-0398, Japan
| | - Isao Shimoyama
- Toyama Prefectural University, Imizu 939-0398, Japan
- The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
10
|
Xu D, Xiao H, Wang S, Li H, Chen HJ, Liu C, Hu N. Universal and Sensitive Drug Assessment Biosensing Platform Using Optimal Mechanical Beating Detection of Single Cardiomyocyte. ACS NANO 2022; 16:15484-15494. [PMID: 36094397 DOI: 10.1021/acsnano.2c08049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The preclinical assessment of efficacy and safety is essential for cardiovascular drug development in order to guarantee effective prevention and treatment of cardiovascular disease and avoid human health endangerment and a huge waste of resources. Rhythmic mechanical beating as one of the crucial cardiomyocyte properties has been exploited to establish a drug assessment biosensing platform. However, the conventional label-free biosensing platforms are difficult to perform high-throughput and high-resolution mechanical beating detection for a single cardiomyocyte, while label-based strategies are limited by pharmacologically adverse effects and phototoxicity. Herein, we propose a biosensing platform involving the multichannel electrode array device and the universal mechanical beating detection system. The platform can determine the optimal characteristic working frequency of different devices and dynamically interrogate the viability of multisite single cardiomyocytes to establish the optimized cell-based model for sensitive drug assessment. The subtle changes of mechanical beating signals induced by cardiovascular drugs can be detected by the platform, thereby demonstrating its high performance in pharmacological assessment. The universal and sensitive drug assessment biosensing platform is believed to be widely applied in cardiology investigating and preclinical drug screening.
Collapse
Affiliation(s)
- Dongxin Xu
- 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
| | - Hongbo Xiao
- 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
| | - Shuzhe Wang
- 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
| | - 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
| | - Hui-Jiuan Chen
- 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
| | - Chuan Liu
- 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
| | - 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
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311200, China
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai 200050, China
| |
Collapse
|
11
|
Nadine S, Chung A, Diltemiz SE, Yasuda B, Lee C, Hosseini V, Karamikamkar S, de Barros NR, Mandal K, Advani S, Zamanian BB, Mecwan M, Zhu Y, Mofidfar M, Zare MR, Mano J, Dokmeci MR, Alambeigi F, Ahadian S. Advances in microfabrication technologies in tissue engineering and regenerative medicine. Artif Organs 2022; 46:E211-E243. [PMID: 35349178 DOI: 10.1111/aor.14232] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tissue engineering provides various strategies to fabricate an appropriate microenvironment to support the repair and regeneration of lost or damaged tissues. In this matter, several technologies have been implemented to construct close-to-native three-dimensional structures at numerous physiological scales, which are essential to confer the functional characteristics of living tissues. METHODS In this article, we review a variety of microfabrication technologies that are currently utilized for several tissue engineering applications, such as soft lithography, microneedles, templated and self-assembly of microstructures, microfluidics, fiber spinning, and bioprinting. RESULTS These technologies have considerably helped us to precisely manipulate cells or cellular constructs for the fabrication of biomimetic tissues and organs. Although currently available tissues still lack some crucial functionalities, including vascular networks, innervation, and lymphatic system, microfabrication strategies are being proposed to overcome these issues. Moreover, the microfabrication techniques that have progressed to the preclinical stage are also discussed. CONCLUSIONS This article aims to highlight the advantages and drawbacks of each technique and areas of further research for a more comprehensive and evolving understanding of microfabrication techniques in terms of tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Sara Nadine
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Ada Chung
- Department of Psychology, University of California-Los Angeles, Los Angeles, California, USA
| | | | - Brooke Yasuda
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,Department of Psychology, University of California-Los Angeles, Los Angeles, California, USA
| | - Charles Lee
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA.,Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, USA.,Station 1, Lawrence, Massachusetts, USA
| | - Vahid Hosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Solmaz Karamikamkar
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | | | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Shailesh Advani
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | | | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Mohammad Mofidfar
- Department of Chemistry, Stanford University, Palo Alto, California, USA
| | | | - João Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, California, USA
| |
Collapse
|
12
|
Dou W, Malhi M, Zhao Q, Wang L, Huang Z, Law J, Liu N, Simmons CA, Maynes JT, Sun Y. Microengineered platforms for characterizing the contractile function of in vitro cardiac models. MICROSYSTEMS & NANOENGINEERING 2022; 8:26. [PMID: 35299653 PMCID: PMC8882466 DOI: 10.1038/s41378-021-00344-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 05/08/2023]
Abstract
Emerging heart-on-a-chip platforms are promising approaches to establish cardiac cell/tissue models in vitro for research on cardiac physiology, disease modeling and drug cardiotoxicity as well as for therapeutic discovery. Challenges still exist in obtaining the complete capability of in situ sensing to fully evaluate the complex functional properties of cardiac cell/tissue models. Changes to contractile strength (contractility) and beating regularity (rhythm) are particularly important to generate accurate, predictive models. Developing new platforms and technologies to assess the contractile functions of in vitro cardiac models is essential to provide information on cell/tissue physiologies, drug-induced inotropic responses, and the mechanisms of cardiac diseases. In this review, we discuss recent advances in biosensing platforms for the measurement of contractile functions of in vitro cardiac models, including single cardiomyocytes, 2D monolayers of cardiomyocytes, and 3D cardiac tissues. The characteristics and performance of current platforms are reviewed in terms of sensing principles, measured parameters, performance, cell sources, cell/tissue model configurations, advantages, and limitations. In addition, we highlight applications of these platforms and relevant discoveries in fundamental investigations, drug testing, and disease modeling. Furthermore, challenges and future outlooks of heart-on-a-chip platforms for in vitro measurement of cardiac functional properties are discussed.
Collapse
Affiliation(s)
- Wenkun Dou
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Manpreet Malhi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Qili Zhao
- Institute of Robotics and Automatic Information System and the Tianjin Key Laboratory of Intelligent Robotics, Nankai University, Tianjin, 300350 China
| | - Li Wang
- School of Mechanical & Automotive Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353 China
| | - Zongjie Huang
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
| | - Na Liu
- School of Mechatronics Engineering and Automation, Shanghai University, Shanghai, 200444 China
| | - Craig A. Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1 Canada
| | - Jason T. Maynes
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON M5S 1A8 Canada
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8 Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9 Canada
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4 Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5T 3A1 Canada
| |
Collapse
|
13
|
Park J, Wu Z, Steiner PR, Zhu B, Zhang JXJ. Heart-on-Chip for Combined Cellular Dynamics Measurements and Computational Modeling Towards Clinical Applications. Ann Biomed Eng 2022; 50:111-137. [PMID: 35039976 DOI: 10.1007/s10439-022-02902-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/01/2022] [Indexed: 12/24/2022]
Abstract
Organ-on-chip or micro-engineered three-dimensional cellular or tissue models are increasingly implemented in the study of cardiovascular pathophysiology as alternatives to traditional in vitro cell culture. Drug induced cardiotoxicity is a key issue in drug development pipelines, but the current in vitro and in vivo studies suffer from inter-species differences, high costs, and lack of reliability and accuracy in predicting cardiotoxicity. Microfluidic heart-on-chip devices can impose a paradigm shift to the current tools. They can not only recapitulate cardiac tissue level functionality and the communication between cells and extracellular matrices but also allow higher throughput studies conducive to drug screening especially with their added functionalities or sensors that extract disease-specific phenotypic, genotypic, and electrophysiological information in real-time. Such electrical and mechanical components can tailor the electrophysiology and mechanobiology of the experiment to better mimic the in vivo condition as well. Recent advancements and challenges are reviewed in the fabrication, functionalization and sensor assisted mechanical and electrophysiological measurements, numerical and computational modeling of cardiomyocytes' behavior, and the clinical applications in drug screening and disease modeling. This review concludes with the current challenges and perspectives on the future of such organ-on-chip platforms.
Collapse
Affiliation(s)
- Jiyoon Park
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Ziqian Wu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Paul R Steiner
- Dartmouth-Hitchcock Medical Center, 1 Medical Center Dr, Lebanon, NH, 03766, USA
| | - Bo Zhu
- Computer Science Department, Dartmouth College, Hanover, NH, 03755, USA
| | - John X J Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA. .,Dartmouth-Hitchcock Medical Center, 1 Medical Center Dr, Lebanon, NH, 03766, USA.
| |
Collapse
|
14
|
Tajabadi M, Goran Orimi H, Ramzgouyan MR, Nemati A, Deravi N, Beheshtizadeh N, Azami M. Regenerative strategies for the consequences of myocardial infarction: Chronological indication and upcoming visions. Biomed Pharmacother 2021; 146:112584. [PMID: 34968921 DOI: 10.1016/j.biopha.2021.112584] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Heart muscle injury and an elevated troponin level signify myocardial infarction (MI), which may result in defective and uncoordinated segments, reduced cardiac output, and ultimately, death. Physicians apply thrombolytic therapy, coronary artery bypass graft (CABG) surgery, or percutaneous coronary intervention (PCI) to recanalize and restore blood flow to the coronary arteries, albeit they were not convincingly able to solve the heart problems. Thus, researchers aim to introduce novel substitutional therapies for regenerating and functionalizing damaged cardiac tissue based on engineering concepts. Cell-based engineering approaches, utilizing biomaterials, gene, drug, growth factor delivery systems, and tissue engineering are the most leading studies in the field of heart regeneration. Also, understanding the primary cause of MI and thus selecting the most efficient treatment method can be enhanced by preparing microdevices so-called heart-on-a-chip. In this regard, microfluidic approaches can be used as diagnostic platforms or drug screening in cardiac disease treatment. Additionally, bioprinting technique with whole organ 3D printing of human heart with major vessels, cardiomyocytes and endothelial cells can be an ideal goal for cardiac tissue engineering and remarkable achievement in near future. Consequently, this review discusses the different aspects, advancements, and challenges of the mentioned methods with presenting the advantages and disadvantages, chronological indications, and application prospects of various novel therapeutic approaches.
Collapse
Affiliation(s)
- Maryam Tajabadi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran
| | - Hanif Goran Orimi
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology (IUST), Narmak, Tehran 16844, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Roya Ramzgouyan
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alireza Nemati
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
15
|
Liao Y, Zhu L, Wang Y. Maturation of Stem Cell-Derived Cardiomyocytes: Foe in Translation Medicine. Int J Stem Cells 2021; 14:366-385. [PMID: 34711701 PMCID: PMC8611306 DOI: 10.15283/ijsc21077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022] Open
Abstract
With the in-depth study of heart development, many human cardiomyocytes (CMs) have been generated in a laboratory environment. CMs derived from pluripotent stem cells (PSCs) have been widely used for a series of applications such as laboratory studies, drug toxicology screening, cardiac disease models, and as an unlimited resource for cell-based cardiac regeneration therapy. However, the low maturity of the induced CMs significantly impedes their applicability. Scientists have been committed to improving the maturation of CMs to achieve the purpose of heart regeneration in the past decades. In this review, we take CMs maturation as the main object of discussion, describe the characteristics of CMs maturation, summarize the key regulatory mechanism of regulating maturation and address the approaches to promote CMs maturation. The maturation of CM is gradually improving due to the incorporation of advanced technologies and is expected to continue.
Collapse
Affiliation(s)
- Yingnan Liao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - Liyuan Zhu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - Yan Wang
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
16
|
Niari SA, Rahbarghazi R, Geranmayeh MH, Karimipour M. Biomaterials patterning regulates neural stem cells fate and behavior: The interface of biology and material science. J Biomed Mater Res A 2021; 110:725-737. [PMID: 34751503 DOI: 10.1002/jbm.a.37321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 11/12/2022]
Abstract
The combination of nanotechnology and stem cell biology is one of the most promising advances in the field of regenerative medicine. This novel combination has widely been utilized in vitro settings in an attempt to develop efficient therapeutic strategies to overcome the limited capacity of the central nervous system (CNS) in replacing degenerating neural cells with functionally normal cells after the onset of acute and chronic neurological disorders. Importantly, biomaterials, not only, enhance the endogenous CNS neurogenesis and plasticity, but also, could provide a desirable supportive microenvironment to harness the full potential of the in vitro expanded neural stem cells (NSCs) for regenerative purposes. Here, first, we discuss how the physical and biochemical properties of biomaterials, such as their stiffness and elasticity, could influence the behavior of NSCs. Then, since the NSCs niche or microenvironment is of fundamental importance in controlling the dynamic destiny of NSCs such as their quiescent and proliferative states, topographical effects of surface diversity in biomaterials, that is, the micro-and nano-patterned surfaces will be discussed in detail. Finally, the influence of biomaterials as artificial microenvironments on the behavior of NSCs through the specific mechanotransduction signaling pathway mediated by focal adhesion formation will be reviewed.
Collapse
Affiliation(s)
- Shabnam Asghari Niari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Geranmayeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Turnbull IC, Zhu W, Stillitano F, Chien CC, Gaitas A. A micromachined force sensing apparatus and method for human engineered cardiac tissue and induced pluripotent stem cell characterization. SENSORS AND ACTUATORS. A, PHYSICAL 2021; 331:112874. [PMID: 34305317 PMCID: PMC8294102 DOI: 10.1016/j.sna.2021.112874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Induced pluripotent stem cell derived-cardiomyocytes (iPSC-CMs) have great potential for cell therapy, drug assessment, and for understanding the pathophysiology and genetic underpinnings of cardiac diseases. Contraction forces are one of the most important characteristics of cardiac function and are predictors of healthy and diseased states. Cantilever techniques, such as atomic force microscopy, measure the vertical force of a single cell, while systems designed to more closely resemble the physical heart function, such as engineered cardiac tissue held by end-posts, measure the axial force. One important question is how do these two force measurements correlate? By establishing a correlation of the axial and vertical force, we will be one step closer in being able to use single cell iPSC-CMs as models. A novel micromachined sensor for measuring force contractions of engineered tissue has been developed. Using this novel sensor, a correlation between axial force and vertical force is experimentally established. This finding supports the use of vertical measurements as an alternative to tissue measurements.
Collapse
Affiliation(s)
| | - Weibin Zhu
- Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | | | - Chen-Chi Chien
- Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Angelo Gaitas
- Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
18
|
Roshanzadeh A, Oyunbaatar NE, Ganjbakhsh SE, Park S, Kim DS, Kanade PP, Lee S, Lee DW, Kim ES. Exposure to nanoplastics impairs collective contractility of neonatal cardiomyocytes under electrical synchronization. Biomaterials 2021; 278:121175. [PMID: 34628193 DOI: 10.1016/j.biomaterials.2021.121175] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/16/2021] [Accepted: 09/30/2021] [Indexed: 10/20/2022]
Abstract
Nanoplastics are global pollutants that have been increasingly released into the environment following the degradation process of industrial and consumer products. These tiny particles have been reported to adversely affect various organs in the body, including the heart. Since it is probable that the less-developed hearts of newborn offspring are more vulnerable to nanoplastic insult during the infant feeding compared with mature hearts of adults, the acute effects of nanoplastics on the collective contractility of neonatal cardiomyocytes are to be elucidated. Here, we traced the aggregation of nanoplastics on the cell membrane and their internalization into the cytosol of neonatal rat ventricular myocytes (NRVMs) for 60 min in the presence of electrical pulses to synchronize the cardiac contraction in vitro. The time-coursed linkage of collective contraction forces, intracellular Ca2+ concentrations, mitochondrial membrane potentials, extracellular field potentials, and reactive oxygen species levels enabled us to build up the sequence of the cellular events associated with the detrimental effects of nanoplastics with positive surface charges on the immature cardiomyocytes. A significant decrease in intracellular Ca2+ levels and electrophysiological activities of NRVMs resulted in the reduction of contraction forces in the early phase (0-15 min). The further reduction of contraction force in the late phase (30-60 min) was attributed to remarkable decreases in mitochondrial membrane potentials and cellular metabolism. Our multifaceted assessments on the effect of positively surface charged nanoplastics on NRVM may offer better understanding of substantial risks of ever-increasing nanoplastic pollution in the hearts of human infants or adults.
Collapse
Affiliation(s)
- Amir Roshanzadeh
- School of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | | | - Sangwoo Park
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju, 61751, Republic of Korea
| | - Dong-Su Kim
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Pooja P Kanade
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49 Dosicheomdansaneopro, Nam-gu, Gwangju, 61751, Republic of Korea
| | - Dong-Weon Lee
- School of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Center for Next Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Eung-Sam Kim
- School of Biological Sciences and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea; Center for Next Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Biological Sciences and Research Center of Ecomimetics, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
19
|
Zhao B, Zhang K, Chen CS, Lejeune E. Sarc-Graph: Automated segmentation, tracking, and analysis of sarcomeres in hiPSC-derived cardiomyocytes. PLoS Comput Biol 2021; 17:e1009443. [PMID: 34613960 PMCID: PMC8523047 DOI: 10.1371/journal.pcbi.1009443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 10/18/2021] [Accepted: 09/10/2021] [Indexed: 12/03/2022] Open
Abstract
A better fundamental understanding of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) has the potential to advance applications ranging from drug discovery to cardiac repair. Automated quantitative analysis of beating hiPSC-CMs is an important and fast developing component of the hiPSC-CM research pipeline. Here we introduce “Sarc-Graph,” a computational framework to segment, track, and analyze sarcomeres in fluorescently tagged hiPSC-CMs. Our framework includes functions to segment z-discs and sarcomeres, track z-discs and sarcomeres in beating cells, and perform automated spatiotemporal analysis and data visualization. In addition to reporting good performance for sarcomere segmentation and tracking with little to no parameter tuning and a short runtime, we introduce two novel analysis approaches. First, we construct spatial graphs where z-discs correspond to nodes and sarcomeres correspond to edges. This makes measuring the network distance between each sarcomere (i.e., the number of connecting sarcomeres separating each sarcomere pair) straightforward. Second, we treat tracked and segmented components as fiducial markers and use them to compute the approximate deformation gradient of the entire tracked population. This represents a new quantitative descriptor of hiPSC-CM function. We showcase and validate our approach with both synthetic and experimental movies of beating hiPSC-CMs. By publishing Sarc-Graph, we aim to make automated quantitative analysis of hiPSC-CM behavior more accessible to the broader research community. Heart disease is the leading cause of death worldwide. Because of this, many researchers are studying heart cells in the lab and trying to create artificial heart tissue. Recently, there has been a growing focus on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). These are cells that are safely sampled from living humans, for example from the blood or skin, that are then transformed into human heart muscle cells. One active research goal is to use these cells to repair the damaged heart. Another active research goal is to test new drugs on these cells before testing them in animals and humans. However, one major challenge is that hiPSC-CMs often have an irregular internal structure that is difficult to analyze. At present, their behavior is far from fully understood. To address this, we have created software to automatically analyze movies of beating hiPSC-CMs. With our software, it is possible to quantify properties such as the amount and direction of beating cell contraction, and the variation in behavior across different parts of each cell. These tools will enable further quantitative analysis of hiPSC-CMs. With these tools, it will be easier to understand, control, and optimize artificial heart tissue created with hiPSC-CMs, and quantify the effects of drugs on hiPSC-CM behavior.
Collapse
Affiliation(s)
- Bill Zhao
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, United States of America
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- The Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, United States of America
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
20
|
Swiatlowska P, Iskratsch T. Tools for studying and modulating (cardiac muscle) cell mechanics and mechanosensing across the scales. Biophys Rev 2021; 13:611-623. [PMID: 34765044 PMCID: PMC8553672 DOI: 10.1007/s12551-021-00837-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
Cardiomyocytes generate force for the contraction of the heart to pump blood into the lungs and body. At the same time, they are exquisitely tuned to the mechanical environment and react to e.g. changes in cell and extracellular matrix stiffness or altered stretching due to reduced ejection fraction in heart disease, by adapting their cytoskeleton, force generation and cell mechanics. Both mechanical sensing and cell mechanical adaptations are multiscale processes. Receptor interactions with the extracellular matrix at the nanoscale will lead to clustering of receptors and modification of the cytoskeleton. This in turn alters mechanosensing, force generation, cell and nuclear stiffness and viscoelasticity at the microscale. Further, this affects cell shape, orientation, maturation and tissue integration at the microscale to macroscale. A variety of tools have been developed and adapted to measure cardiomyocyte receptor-ligand interactions and forces or mechanics at the different ranges, resulting in a wealth of new information about cardiomyocyte mechanobiology. Here, we take stock at the different tools for exploring cardiomyocyte mechanosensing and cell mechanics at the different scales from the nanoscale to microscale and macroscale.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
21
|
He C, Wei X, Liang T, Liu M, Jiang D, Zhuang L, Wang P. Quantifying the Compressive Force of 3D Cardiac Tissues via Calculating the Volumetric Deformation of Built-In Elastic Gelatin Microspheres. Adv Healthc Mater 2021; 10:e2001716. [PMID: 34197053 DOI: 10.1002/adhm.202001716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/30/2020] [Indexed: 01/28/2023]
Abstract
Quantifying cardiac contractile force is of paramount important in studying mechanical heart failure and screening therapeutic drugs. However, most existing methods can only measure the in-plane component of twitch force of cardiomyocytes, such that mismatching the centripetal compressive stress of heart beating in physiology. Here, a non-destructive method is developed for quantifying the compressive stress and mapping the distribution of the local stress within the 3D cardiac tissues. In detail, elastic gelatin microspheres labeled with fluorescence beads are fabricated by microfluidic chips with high throughput, and they serve as built-in pressure sensors which are wrapped by cardiomyocytes in 3D tissues. The deformation of microspheres and the displacements of fluorescent beads induced by the contraction of cardiomyocytes are demonstrated to characterize the amount and distribution of the centripetal compressive stress. Further, the method shows a potent capability to locally quantify contractile force variation of 3D cardiac tissues, which is induced by agonist (norepinephrine) and inhibitor (blebbistatin). On the whole, the method significantly improves the 3D measurement of mechanical force in vitro and provides a solution for locally quantifying the compressive stress within engineered cardiac tissues.
Collapse
Affiliation(s)
- Chuanjiang He
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| | - Xinwei Wei
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Tao Liang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Mengxue Liu
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Deming Jiang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Liujing Zhuang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
| | - Ping Wang
- Biosensor National Special Laboratory Key Laboratory for Biomedical Engineering Ministry of Education Department of Biomedical Engineering Zhejiang University Hangzhou 310027 China
- State Key Laboratory of Transducer Technology Chinese Academy of Sciences Shanghai 200050 China
| |
Collapse
|
22
|
Andrysiak K, Stępniewski J, Dulak J. Human-induced pluripotent stem cell-derived cardiomyocytes, 3D cardiac structures, and heart-on-a-chip as tools for drug research. Pflugers Arch 2021; 473:1061-1085. [PMID: 33629131 PMCID: PMC8245367 DOI: 10.1007/s00424-021-02536-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/13/2022]
Abstract
Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.
Collapse
Affiliation(s)
- Kalina Andrysiak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
23
|
Müller D, Klamt T, Gentemann L, Heisterkamp A, Kalies SMK. Evaluation of laser induced sarcomere micro-damage: Role of damage extent and location in cardiomyocytes. PLoS One 2021; 16:e0252346. [PMID: 34086732 PMCID: PMC8177425 DOI: 10.1371/journal.pone.0252346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
Whereas it is evident that a well aligned and regular sarcomeric structure in cardiomyocytes is vital for heart function, considerably less is known about the contribution of individual elements to the mechanics of the entire cell. For instance, it is unclear whether altered Z-disc elements are the reason or the outcome of related cardiomyopathies. Therefore, it is crucial to gain more insight into this cellular organization. This study utilizes femtosecond laser-based nanosurgery to better understand sarcomeres and their repair upon damage. We investigated the influence of the extent and the location of the Z-disc damage. A single, three, five or ten Z-disc ablations were performed in neonatal rat cardiomyocytes. We employed image-based analysis using a self-written software together with different already published algorithms. We observed that cardiomyocyte survival associated with the damage extent, but not with the cell area or the total number of Z-discs per cell. The cell survival is independent of the damage position and can be compensated. However, the sarcomere alignment/orientation is changing over time after ablation. The contraction time is also independent of the extent of damage for the tested parameters. Additionally, we observed shortening rates between 6–7% of the initial sarcomere length in laser treated cardiomyocytes. This rate is an important indicator for force generation in myocytes. In conclusion, femtosecond laser-based nanosurgery together with image-based sarcomere tracking is a powerful tool to better understand the Z-disc complex and its force propagation function and role in cellular mechanisms.
Collapse
Affiliation(s)
- Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- * E-mail:
| | - Thorben Klamt
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Stefan Michael Klaus Kalies
- Institute of Quantum Optics, Leibniz University Hannover, Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
24
|
Tsui JH, Leonard A, Camp ND, Long JT, Nawas ZY, Chavanachat R, Smith AST, Choi JS, Dong Z, Ahn EH, Wolf-Yadlin A, Murry CE, Sniadecki NJ, Kim DH. Tunable electroconductive decellularized extracellular matrix hydrogels for engineering human cardiac microphysiological systems. Biomaterials 2021; 272:120764. [PMID: 33798964 DOI: 10.1016/j.biomaterials.2021.120764] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes differentiated from human induced pluripotent stem cells (hiPSCs) offer tremendous potential when used to engineer human tissues for drug screening and disease modeling; however, phenotypic immaturity reduces assay reliability when translating in vitro results to clinical studies. To address this, we have developed hybrid hydrogels comprised of decellularized porcine myocardial extracellular matrix (dECM) and reduced graphene oxide (rGO) to provide a more instructive microenvironment for proper cell and tissue development. A tissue-specific protein profile was preserved post-decellularization, and through the modulation of rGO content and degree of reduction, the mechanical and electrical properties of the hydrogels could be tuned. Engineered heart tissues (EHTs) generated using dECM-rGO hydrogel scaffolds and hiPSC-derived cardiomyocytes exhibited significantly increased twitch forces and had increased expression of genes that regulate contractile function. Improvements in various aspects of electrophysiological function, such as calcium-handling, action potential duration, and conduction velocity, were also induced by the hybrid biomaterial. dECM-rGO hydrogels could also be used as a bioink to print cardiac tissues in a high-throughput manner, and these tissues were utilized to assess the proarrhythmic potential of cisapride. Action potential prolongation and beat interval irregularities was observed in dECM-rGO tissues at clinical doses of cisapride, indicating that the enhanced electrophysiological function of these tissues corresponded well with a capability to produce physiologically relevant drug responses.
Collapse
Affiliation(s)
- Jonathan H Tsui
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Andrea Leonard
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA
| | - Nathan D Camp
- Department of Genome Sciences, University of Washington, Seattle, WA, 98105, USA
| | - Joseph T Long
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | - Zeid Y Nawas
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA
| | | | - Alec S T Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, 98105, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Jong Seob Choi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Zhipeng Dong
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Charles E Murry
- Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; 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 & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Nathan J Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98105, USA; Department of Bioengineering, University of Washington, Seattle, WA, 98105, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA; Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA; Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
House A, Atalla I, Lee EJ, Guvendiren M. Designing Biomaterial Platforms for Cardiac Tissue and Disease Modeling. ADVANCED NANOBIOMED RESEARCH 2021; 1:2000022. [PMID: 33709087 PMCID: PMC7942203 DOI: 10.1002/anbr.202000022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart disease is one of the leading causes of death in the world. There is a growing demand for in vitro cardiac models that can recapitulate the complex physiology of the cardiac tissue. These cardiac models can provide a platform to better understand the underlying mechanisms of cardiac development and disease and aid in developing novel treatment alternatives and platforms towards personalized medicine. In this review, a summary of engineered cardiac platforms is presented. Basic design considerations for replicating the heart's microenvironment are discussed considering the anatomy of the heart. This is followed by a detailed summary of the currently available biomaterial platforms for modeling the heart tissue in vitro. These in vitro models include 2D surface modified structures, 3D molded structures, porous scaffolds, electrospun scaffolds, bioprinted structures, and heart-on-a-chip devices. The challenges faced by current models and the future directions of in vitro cardiac models are also discussed. Engineered in vitro tissue models utilizing patients' own cells could potentially revolutionize the way we develop treatment and diagnostic alternatives.
Collapse
Affiliation(s)
- Andrew House
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Iren Atalla
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Eun Jung Lee
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory, Otto H. York Chemical and Materials Engineering, 138 York Center, University Heights, Newark, NJ 07102, USA
| |
Collapse
|
26
|
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
|
27
|
Cho KW, Lee WH, Kim BS, Kim DH. Sensors in heart-on-a-chip: A review on recent progress. Talanta 2020; 219:121269. [DOI: 10.1016/j.talanta.2020.121269] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/14/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
|
28
|
Mustafa A, Eser A, Aksu AC, Kiraz A, Tanyeri M, Erten A, Yalcin O. A micropillar-based microfluidic viscometer for Newtonian and non-Newtonian fluids. Anal Chim Acta 2020; 1135:107-115. [PMID: 33070846 DOI: 10.1016/j.aca.2020.07.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/30/2020] [Accepted: 07/15/2020] [Indexed: 11/17/2022]
Abstract
In this study, a novel viscosity measurement technique based on measuring the deflection of flexible (poly) dimethylsiloxane (PDMS) micropillars is presented. The experimental results show a nonlinear relationship between fluid viscosity and the deflection of micropillars due to viscoelastic properties of PDMS. A calibration curve, demonstrating this nonlinear relationship, is generated, and used to determine the viscosity of an unknown fluid. Using our method, viscosity measurements for Newtonian fluids (glycerol/water solutions) can be performed within 2-100 cP at shear rates γ = 60.5-398.4 s-1. We also measured viscosity of human whole blood samples (non-Newtonian fluid) yielding 2.7-5.1 cP at shear rates γ = 120-345.1 s-1, which compares well with measurements using conventional rotational viscometers (3.6-5.7 cP). With a sensitivity better than 0.5 cP, this method has the potential to be used as a portable microfluidic viscometer for real-time rheological studies.
Collapse
Affiliation(s)
- Adil Mustafa
- Graduate School of Biomedical Sciences and Engineering Koç University, Istanbul, Turkey; Department of Physics, Koç University, Istanbul, Turkey
| | - Aysenur Eser
- Graduate School of Biomedical Sciences and Engineering Koç University, Istanbul, Turkey
| | - Ali Cenk Aksu
- Graduate School of Biomedical Sciences and Engineering Koç University, Istanbul, Turkey
| | - Alper Kiraz
- Department of Physics, Koç University, Istanbul, Turkey; Department of Electrical Engineering Koç University, Istanbul, Turkey.
| | - Melikhan Tanyeri
- Department of Engineering, Duquesne University, Pittsburgh, USA.
| | - Ahmet Erten
- Department of Electronics and Communication Engineering, Istanbul Technical University, Istanbul, Turkey.
| | - Ozlem Yalcin
- Research Center for Translational Medicine, School of Medicine, Koç University, Istanbul, Turkey.
| |
Collapse
|
29
|
Obal D, Wu JC. Induced pluripotent stem cells as a platform to understand patient-specific responses to opioids and anaesthetics. Br J Pharmacol 2020; 177:4581-4594. [PMID: 32767563 PMCID: PMC7520445 DOI: 10.1111/bph.15228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/22/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
Recent advances in human induced pluripotent stem cell (iPSC) technology may provide unprecedented opportunities to study patient-specific responses to anaesthetics and opioids. In this review, we will (1) examine the advantages and limitations of iPSC technology, (2) summarize studies using iPSCs that have contributed to our current understanding of anaesthetics and opioid action on the cardiovascular system and central nervous system (CNS), and (3) describe how iPSC technology can be used to further develop personalized analgesic and sedative pharmacotherapies with reduced or minimal detrimental cardiovascular effects.
Collapse
Affiliation(s)
- Detlef Obal
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Anesthesiology, Pain, and Perioperative MedicineStanford UniversityStanfordCaliforniaUSA
- Outcomes Research ConsortiumClevelandOhioUSA
| | - Joseph C. Wu
- Stanford Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of Medicine, Division of Cardiovascular MedicineStanford UniversityStanfordCaliforniaUSA
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
30
|
Huang H, Dai C, Shen H, Gu M, Wang Y, Liu J, Chen L, Sun L. Recent Advances on the Model, Measurement Technique, and Application of Single Cell Mechanics. Int J Mol Sci 2020; 21:E6248. [PMID: 32872378 PMCID: PMC7504142 DOI: 10.3390/ijms21176248] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since the cell was discovered by humans, it has been an important research subject for researchers. The mechanical response of cells to external stimuli and the biomechanical response inside cells are of great significance for maintaining the life activities of cells. These biomechanical behaviors have wide applications in the fields of disease research and micromanipulation. In order to study the mechanical behavior of single cells, various cell mechanics models have been proposed. In addition, the measurement technologies of single cells have been greatly developed. These models, combined with experimental techniques, can effectively explain the biomechanical behavior and reaction mechanism of cells. In this review, we first introduce the basic concept and biomechanical background of cells, then summarize the research progress of internal force models and experimental techniques in the field of cell mechanics and discuss the latest mechanical models and experimental methods. We summarize the application directions of cell mechanics and put forward the future perspectives of a cell mechanics model.
Collapse
Affiliation(s)
| | | | | | | | | | - Jizhu Liu
- School of Mechanical and Electric Engineering, Jiangsu Provincial Key Laboratory of Advanced Robotics, Soochow University, Suzhou 215123, China; (H.H.); (C.D.); (H.S.); (M.G.); (Y.W.); (L.S.)
| | - Liguo Chen
- School of Mechanical and Electric Engineering, Jiangsu Provincial Key Laboratory of Advanced Robotics, Soochow University, Suzhou 215123, China; (H.H.); (C.D.); (H.S.); (M.G.); (Y.W.); (L.S.)
| | | |
Collapse
|
31
|
Yokota T, McCourt J, Ma F, Ren S, Li S, Kim TH, Kurmangaliyev YZ, Nasiri R, Ahadian S, Nguyen T, Tan XHM, Zhou Y, Wu R, Rodriguez A, Cohn W, Wang Y, Whitelegge J, Ryazantsev S, Khademhosseini A, Teitell MA, Chiou PY, Birk DE, Rowat AC, Crosbie RH, Pellegrini M, Seldin M, Lusis AJ, Deb A. Type V Collagen in Scar Tissue Regulates the Size of Scar after Heart Injury. Cell 2020; 182:545-562.e23. [PMID: 32621799 PMCID: PMC7415659 DOI: 10.1016/j.cell.2020.06.030] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/17/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.
Collapse
Affiliation(s)
- Tomohiro Yokota
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Jackie McCourt
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Shuxun Ren
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Tae-Hyung Kim
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Yerbol Z Kurmangaliyev
- Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Rohollah Nasiri
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Samad Ahadian
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA
| | - Thang Nguyen
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA
| | - Xing Haw Marvin Tan
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | - Yonggang Zhou
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Rimao Wu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Abraham Rodriguez
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Whitaker Cohn
- Passarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience and Behaviour, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Julian Whitelegge
- Passarow Mass Spectrometry Laboratory, Semel Institute for Neuroscience and Behaviour, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Sergey Ryazantsev
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Ali Khademhosseini
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90024, USA; Department of Chemical Engineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Radiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Michael A Teitell
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Pei-Yu Chiou
- California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA; Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
| | - David E Birk
- University of South Florida College of Medicine, Tampa, FL 33612, USA
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, University of California, CA 90095, USA; Department of Bioengineering, School of Engineering, University of California, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Marcus Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Genetics, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; UCLA Cardiovascular Theme, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California, Los Angeles, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; California Nanosystems Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Lysosomal Abnormalities in Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21030811. [PMID: 32012649 PMCID: PMC7036830 DOI: 10.3390/ijms21030811] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/25/2020] [Indexed: 12/12/2022] Open
Abstract
The lysosome, a key organelle for cellular clearance, is associated with a wide variety of pathological conditions in humans. Lysosome function and its related pathways are particularly important for maintaining the health of the cardiovascular system. In this review, we highlighted studies that have improved our understanding of the connection between lysosome function and cardiovascular diseases with an emphasis on a recent breakthrough that characterized a unique autophagosome-lysosome fusion mechanism employed by cardiomyocytes through a lysosomal membrane protein LAMP-2B. This finding may impact the development of future therapeutic applications.
Collapse
|
35
|
|
36
|
Manini P, Lucci V, Lino V, Sartini S, Rossella F, Falco G, Chiappe C, d’Ischia M. Synthetic mycomelanin thin films as emergent bio-inspired interfaces controlling the fate of embryonic stem cells. J Mater Chem B 2020; 8:4412-4418. [DOI: 10.1039/d0tb00623h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mycomelanin thin films from 1,8-dihydroxynaphthalene can serve as a biointerface inducing adhesion and proliferation of ESCs and promoting their differentiation towards endodermal lineages.
Collapse
Affiliation(s)
- Paola Manini
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Valeria Lucci
- Department of Biology
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Valeria Lino
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | | | - Francesco Rossella
- NEST
- Scuola Normale Superiore and Istituto Nanoscienze – CNR
- I-56127 Pisa
- Italy
| | - Geppino Falco
- Department of Biology
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| | - Cinzia Chiappe
- Department of Pharmacy
- University of Pisa
- I-56126 Pisa
- Italy
| | - Marco d’Ischia
- Department of Chemical Sciences
- University of Napoli Federico II
- Complesso Universitario Monte S. Angelo
- I-80126 Napoli
- Italy
| |
Collapse
|
37
|
Miklas JW, Clark E, Levy S, Detraux D, Leonard A, Beussman K, Showalter MR, Smith AT, Hofsteen P, Yang X, Macadangdang J, Manninen T, Raftery D, Madan A, Suomalainen A, Kim DH, Murry CE, Fiehn O, Sniadecki NJ, Wang Y, Ruohola-Baker H. TFPa/HADHA is required for fatty acid beta-oxidation and cardiolipin re-modeling in human cardiomyocytes. Nat Commun 2019; 10:4671. [PMID: 31604922 PMCID: PMC6789043 DOI: 10.1038/s41467-019-12482-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial trifunctional protein deficiency, due to mutations in hydratase subunit A (HADHA), results in sudden infant death syndrome with no cure. To reveal the disease etiology, we generated stem cell-derived cardiomyocytes from HADHA-deficient hiPSCs and accelerated their maturation via an engineered microRNA maturation cocktail that upregulated the epigenetic regulator, HOPX. Here we report, matured HADHA mutant cardiomyocytes treated with an endogenous mixture of fatty acids manifest the disease phenotype: defective calcium dynamics and repolarization kinetics which results in a pro-arrhythmic state. Single cell RNA-seq reveals a cardiomyocyte developmental intermediate, based on metabolic gene expression. This intermediate gives rise to mature-like cardiomyocytes in control cells but, mutant cells transition to a pathological state with reduced fatty acid beta-oxidation, reduced mitochondrial proton gradient, disrupted cristae structure and defective cardiolipin remodeling. This study reveals that HADHA (tri-functional protein alpha), a monolysocardiolipin acyltransferase-like enzyme, is required for fatty acid beta-oxidation and cardiolipin remodeling, essential for functional mitochondria in human cardiomyocytes.
Collapse
Affiliation(s)
- Jason W Miklas
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Elisa Clark
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Shiri Levy
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Damien Detraux
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA
| | - Andrea Leonard
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Kevin Beussman
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
| | - Megan R Showalter
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
| | - Alec T Smith
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Peter Hofsteen
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Jesse Macadangdang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Tuula Manninen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Anup Madan
- Covance Genomics Laboratory, Redmond, WA, 98052, USA
| | - Anu Suomalainen
- Helsinki University Hospital, 00290, Helsinki, Finland
- Research Programs Unit, Stem Cells and Metabolism, University of Helsinki, 00290, Helsinki, Finland
- Neuroscience Center, University of Helsinki, 00290, Helsinki, Finland
| | - Deok-Ho Kim
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, 98109, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
- Department of Medicine/Cardiology, University of Washington, Seattle, WA, 98109, USA
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nathan J Sniadecki
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, 98195, USA
- Department of Pathology, University of Washington, Seattle, WA, 98109, USA
| | - Yuliang Wang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Hannele Ruohola-Baker
- Institute for Stem Cell and Regenerative Medicine, University of Washington, School of Medicine, Seattle, WA, 98109, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
- Department of Biochemistry, University of Washington, School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
38
|
Zhou Z, Yu P, Zhou L, Tu L, Fan L, Zhang F, Dai C, Liu Y, Ning C, Du J, Tan G. Polypyrrole Nanocones and Dynamic Piezoelectric Stimulation-Induced Stem Cell Osteogenic Differentiation. ACS Biomater Sci Eng 2019; 5:4386-4392. [PMID: 33438404 DOI: 10.1021/acsbiomaterials.9b00812] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Imitating the physiological microenvironment of living cell and tissues opens new avenues of research into the application of electricity to medical therapies. In this study, dynamic piezoelectric stimulation is generated in a dynamic culture because of the piezoelectric effect of the poly(vinylidene fluoride)-polypyrrole (PVDF-PPy) electroactive composite. Combined with PPy nanocones, dynamic piezoelectric signals are effectively and continuously provided to cells. In the presence of dynamic piezoelectric stimulation and PPy nanocones, PPy-PVDF NS samples show promoted bone mesenchymal stem cell (BMSCs) adhesion, spreadin, and osteogenic differentiation. On the basis of the results of this study, PPy nanocones and dynamic piezoelectric stimulation can be administered to modulate cell behavior, paving the way for the exploration of cellular responses to dynamic electrical stimulation.
Collapse
Affiliation(s)
- Zhengnan Zhou
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Peng Yu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China.,Guangdong Key Laboratory of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lei Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China.,Guangdong Key Laboratory of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Lingjie Tu
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Lei Fan
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China.,Guangdong Key Laboratory of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Fengmiao Zhang
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Cong Dai
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Yi Liu
- Orthopedics Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Chengyun Ning
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China.,Guangdong Key Laboratory of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jianqiang Du
- Department of Nuclear Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Guoxin Tan
- Institute of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
39
|
Affiliation(s)
| | | | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA
| |
Collapse
|
40
|
Callaghan NI, Hadipour-Lakmehsari S, Lee SH, Gramolini AO, Simmons CA. Modeling cardiac complexity: Advancements in myocardial models and analytical techniques for physiological investigation and therapeutic development in vitro. APL Bioeng 2019; 3:011501. [PMID: 31069331 PMCID: PMC6481739 DOI: 10.1063/1.5055873] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathies, heart failure, and arrhythmias or conduction blockages impact millions of patients worldwide and are associated with marked increases in sudden cardiac death, decline in the quality of life, and the induction of secondary pathologies. These pathologies stem from dysfunction in the contractile or conductive properties of the cardiomyocyte, which as a result is a focus of fundamental investigation, drug discovery and therapeutic development, and tissue engineering. All of these foci require in vitro myocardial models and experimental techniques to probe the physiological functions of the cardiomyocyte. In this review, we provide a detailed exploration of different cell models, disease modeling strategies, and tissue constructs used from basic to translational research. Furthermore, we highlight recent advancements in imaging, electrophysiology, metabolic measurements, and mechanical and contractile characterization modalities that are advancing our understanding of cardiomyocyte physiology. With this review, we aim to both provide a biological framework for engineers contributing to the field and demonstrate the technical basis and limitations underlying physiological measurement modalities for biologists attempting to take advantage of these state-of-the-art techniques.
Collapse
Affiliation(s)
| | | | | | | | - Craig A. Simmons
- Author to whom correspondence should be addressed: . Present address: Ted Rogers Centre for Heart
Research, 661 University Avenue, 14th Floor Toronto, Ontario M5G 1M1, Canada. Tel.:
416-946-0548. Fax: 416-978-7753
| |
Collapse
|
41
|
Nandi S, Sproul EP, Nellenbach K, Erb M, Gaffney L, Freytes DO, Brown AC. Platelet-like particles dynamically stiffen fibrin matrices and improve wound healing outcomes. Biomater Sci 2019; 7:669-682. [PMID: 30608063 PMCID: PMC6385160 DOI: 10.1039/c8bm01201f] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Native platelets perform several critical functions within the context of wound healing, including participating in initial hemostasis and interacting with fibrin at the wound site to induce clot retraction. Platelet depletion or dysfunction due to trauma or disease can inhibit robust wound healing responses. There has been a focus recently on developing synthetic, non-immunogenic platelet mimetic technologies for the purpose of augmenting hemostatic responses in cases of deficient native platelet functionality. Here we describe the application of synthetic platelet-like particles (PLPs), capable of recapitulating the deformable platelet body and fibrin specificity found in native platelets, to enhance healing outcomes. We first demonstrate PLPs mimic activated platelet morphology and induce fibrin clot retraction. During clot retraction, native platelets generate forces within a fibrin network to stiffen the fibrin matrix; therefore, we hypothesized that our PLPs will likewise be able to stiffen provisional fibrin matrices. Due to previous studies indicating that increased matrix stiffness is linked to increased cellular migration, we further hypothesize that PLP-mediated fibrin stiffening will enhance cell migration and improve healing outcomes within in vitro and in vivo models of wound healing. PLPs were found to enhance fibroblast migration in in vitro models of early wound healing and enhance healing outcomes in an in vivo murine model of wound healing. These studies demonstrate the utility of PLPs for enhancing wound repair and also provide insight into the role of native platelet-mediated clot retraction in wound healing.
Collapse
Affiliation(s)
- Seema Nandi
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA.
| | | | | | | | | | | | | |
Collapse
|
42
|
LAMP-2B regulates human cardiomyocyte function by mediating autophagosome-lysosome fusion. Proc Natl Acad Sci U S A 2018; 116:556-565. [PMID: 30584088 DOI: 10.1073/pnas.1808618116] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mutations in lysosomal-associated membrane protein 2 (LAMP-2) gene are associated with Danon disease, which often leads to cardiomyopathy/heart failure through poorly defined mechanisms. Here, we identify the LAMP-2 isoform B (LAMP-2B) as required for autophagosome-lysosome fusion in human cardiomyocytes (CMs). Remarkably, LAMP-2B functions independently of syntaxin 17 (STX17), a protein that is essential for autophagosome-lysosome fusion in non-CMs. Instead, LAMP-2B interacts with autophagy related 14 (ATG14) and vesicle-associated membrane protein 8 (VAMP8) through its C-terminal coiled coil domain (CCD) to promote autophagic fusion. CMs derived from induced pluripotent stem cells (hiPSC-CMs) from Danon patients exhibit decreased colocalization between ATG14 and VAMP8, profound defects in autophagic fusion, as well as mitochondrial and contractile abnormalities. This phenotype was recapitulated by LAMP-2B knockout in non-Danon hiPSC-CMs. Finally, gene correction of LAMP-2 mutation rescues the Danon phenotype. These findings reveal a STX17-independent autophagic fusion mechanism in human CMs, providing an explanation for cardiomyopathy in Danon patients and a foundation for targeting defective LAMP-2B-mediated autophagy to treat this patient population.
Collapse
|
43
|
Bruyneel AA, McKeithan WL, Feyen DA, Mercola M. Will iPSC-cardiomyocytes revolutionize the discovery of drugs for heart disease? Curr Opin Pharmacol 2018; 42:55-61. [PMID: 30081259 DOI: 10.1016/j.coph.2018.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/01/2018] [Indexed: 12/30/2022]
Abstract
Cardiovascular disease remains the largest single cause of mortality in the Western world, despite significant advances in clinical management over the years. Unfortunately, the development of new cardiovascular medicines is stagnating and can in part be attributed to the difficulty of screening for novel therapeutic strategies due to a lack of suitable models. The advent of human induced pluripotent stem cells and the ability to make limitless numbers of cardiomyocytes could revolutionize heart disease modeling and drug discovery. This review summarizes the state of the art in the field, describes the strengths and weaknesses of the technology, and applications where the model system would be most appropriate.
Collapse
Affiliation(s)
- Arne An Bruyneel
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wesley L McKeithan
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dries Am Feyen
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Mercola
- The Cardiovascular Institute and Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
44
|
Wang L, Dou W, Malhi M, Zhu M, Liu H, Plakhotnik J, Xu Z, Zhao Q, Chen J, Chen S, Hamilton R, Simmons CA, Maynes JT, Sun Y. Microdevice Platform for Continuous Measurement of Contractility, Beating Rate, and Beating Rhythm of Human-Induced Pluripotent Stem Cell-Cardiomyocytes inside a Controlled Incubator Environment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21173-21183. [PMID: 29874032 DOI: 10.1021/acsami.8b05407] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The heart completes a complex set of tasks, including the initiation or propagation of an electrical signal with regularity (proper heart rate and rhythm) and generating sufficient force of contraction (contractility). Probing mechanisms of heart diseases and quantifying drug efficacies demand a platform that is capable of continuous operation inside a cell incubator for long-term measurement of cardiomyocyte (CM) monolayers. Here, we report a microdevice array that is capable of performing continuous, long-term (14 days) measurement of contractility, beating rate, and beating rhythm in a monolayer of human-induced pluripotent stem cell-CMs (hiPSC-CMs). The device consists of a deformable membrane with embedded carbon nanotube (CNT)-based strain sensors. Contraction of the hiPSC-CMs seeded on the membrane induces electrical resistance change of the CNT strain sensor. Continuously reading the sensor signals revealed that hiPSC-CMs started to beat from day 2 and plateaued on day 5. Average contractile stress generated by a monolayer of hiPSC-CMs was determined to be 2.34 ± 0.041 kPa with a beating rate of 1.17 ± 0.068 Hz. The device arrays were also used to perform comprehensive measurement of the beating rate, rhythm, and contractility of the hiPSC-CMs and quantify the cell responses to different concentrations of agonists and antagonists, which altered the average contractile stress to the range of 1.15 ± 0.13 to 3.96 ± 0.53 kPa. The continuous measurement capability of the device arrays also enabled the generation of Poincaré plots for revealing subtle changes in the beating rhythm of hiPSC-CMs under different drug treatments.
Collapse
Affiliation(s)
- Li Wang
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Wenkun Dou
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Manpreet Malhi
- Hospital for Sick Children , Toronto ON M5G 1X8 , Canada
| | - Min Zhu
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Haijiao Liu
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto ON M5S 3G9 , Canada
| | | | - Zhensong Xu
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Qili Zhao
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Jun Chen
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | - Siyu Chen
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
| | | | - Craig A Simmons
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto ON M5S 3G9 , Canada
| | - Jason T Maynes
- Hospital for Sick Children , Toronto ON M5G 1X8 , Canada
| | - Yu Sun
- Department of Mechanical and Industrial Engineering , University of Toronto , Toronto ON M5S 3G8 , Canada
- Institute of Biomaterials and Biomedical Engineering , University of Toronto , Toronto ON M5S 3G9 , Canada
- Department of Electrical and Computer Engineering , University of Toronto , Toronto ON M5S 3G4 , Canada
| |
Collapse
|
45
|
Chung HH, Mireles M, Kwarta BJ, Gaborski TR. Use of porous membranes in tissue barrier and co-culture models. LAB ON A CHIP 2018; 18:1671-1689. [PMID: 29845145 PMCID: PMC5997570 DOI: 10.1039/c7lc01248a] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Porous membranes enable the partitioning of cellular microenvironments in vitro, while still allowing physical and biochemical crosstalk between cells, a feature that is often necessary for recapitulating physiological functions. This article provides an overview of the different membranes used in tissue barrier and cellular co-culture models with a focus on experimental design and control of these systems. Specifically, we discuss how the structural, mechanical, chemical, and even the optical and transport properties of different membranes bestow specific advantages and disadvantages through the context of physiological relevance. This review also explores how membrane pore properties affect perfusion and solute permeability by developing an analytical framework to guide the design and use of tissue barrier or co-culture models. Ultimately, this review offers insight into the important aspects one must consider when using porous membranes in tissue barrier and lab-on-a-chip applications.
Collapse
Affiliation(s)
- Henry H Chung
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA.
| | | | | | | |
Collapse
|
46
|
Bruyneel AAN, McKeithan WL, Feyen DAM, Mercola M. Using iPSC Models to Probe Regulation of Cardiac Ion Channel Function. Curr Cardiol Rep 2018; 20:57. [DOI: 10.1007/s11886-018-1000-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Wheelwright M, Win Z, Mikkila JL, Amen KY, Alford PW, Metzger JM. Investigation of human iPSC-derived cardiac myocyte functional maturation by single cell traction force microscopy. PLoS One 2018; 13:e0194909. [PMID: 29617427 PMCID: PMC5884520 DOI: 10.1371/journal.pone.0194909] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/13/2018] [Indexed: 11/24/2022] Open
Abstract
Recent advances have made it possible to readily derive cardiac myocytes from human induced pluripotent stem cells (hiPSC-CMs). HiPSC-CMs represent a valuable new experimental model for studying human cardiac muscle physiology and disease. Many laboratories have devoted substantial effort to examining the functional properties of isolated hiPSC-CMs, but to date, force production has not been adequately characterized. Here, we utilized traction force microscopy (TFM) with micro-patterning cell printing to investigate the maximum force production of isolated single hiPSC-CMs under varied culture and assay conditions. We examined the role of length of differentiation in culture and the effects of varied extracellular calcium concentration in the culture media on the maturation of hiPSC-CMs. Results show that hiPSC-CMs developing in culture for two weeks produced significantly less force than cells cultured from one to three months, with hiPSC-CMs cultured for three months resembling the cell morphology and function of neonatal rat ventricular myocytes in terms of size, dimensions, and force production. Furthermore, hiPSC-CMs cultured long term in conditions of physiologic calcium concentrations were larger and produced more force than hiPSC-CMs cultured in standard media with sub-physiological calcium. We also examined relationships between cell morphology, substrate stiffness and force production. Results showed a significant relationship between cell area and force. Implementing directed modifications of substrate stiffness, by varying stiffness from embryonic-like to adult myocardium-like, hiPSC-CMs produced maximal forces on substrates with a lower modulus and significantly less force when assayed on increasingly stiff adult myocardium-like substrates. Calculated strain energy measurements paralleled these findings. Collectively, these findings further establish single cell TFM as a valuable approach to illuminate the quantitative physiological maturation of force in hiPSC-CMs.
Collapse
Affiliation(s)
- Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zaw Win
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jennifer L. Mikkila
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Kamilah Y. Amen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Patrick W. Alford
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
48
|
Broders-Bondon F, Nguyen Ho-Bouldoires TH, Fernandez-Sanchez ME, Farge E. Mechanotransduction in tumor progression: The dark side of the force. J Cell Biol 2018; 217:1571-1587. [PMID: 29467174 PMCID: PMC5940296 DOI: 10.1083/jcb.201701039] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Broders-Bondon et al. review the pathological mechanical properties of tumor tissues and how abnormal mechanical signals result in oncogenic biochemical signals during tumor progression. Cancer has been characterized as a genetic disease, associated with mutations that cause pathological alterations of the cell cycle, adhesion, or invasive motility. Recently, the importance of the anomalous mechanical properties of tumor tissues, which activate tumorigenic biochemical pathways, has become apparent. This mechanical induction in tumors appears to consist of the destabilization of adult tissue homeostasis as a result of the reactivation of embryonic developmental mechanosensitive pathways in response to pathological mechanical strains. These strains occur in many forms, for example, hypervascularization in late tumors leads to high static hydrodynamic pressure that can promote malignant progression through hypoxia or anomalous interstitial liquid and blood flow. The high stiffness of tumors directly induces the mechanical activation of biochemical pathways enhancing the cell cycle, epithelial–mesenchymal transition, and cell motility. Furthermore, increases in solid-stress pressure associated with cell hyperproliferation activate tumorigenic pathways in the healthy epithelial cells compressed by the neighboring tumor. The underlying molecular mechanisms of the translation of a mechanical signal into a tumor inducing biochemical signal are based on mechanically induced protein conformational changes that activate classical tumorigenic signaling pathways. Understanding these mechanisms will be important for the development of innovative treatments to target such mechanical anomalies in cancer.
Collapse
Affiliation(s)
- Florence Broders-Bondon
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Thanh Huong Nguyen Ho-Bouldoires
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Maria-Elena Fernandez-Sanchez
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic and Tumor Development Group, Institut Curie, PSL Research University, Centre National de la Recherche Scientifique, UMR168, Inserm, Sorbonne Universities, Paris, France
| |
Collapse
|
49
|
Pushkarsky I, Tseng P, Black D, France B, Warfe L, Koziol-White CJ, Jester WF, Trinh RK, Lin J, Scumpia PO, Morrison SL, Panettieri RA, Damoiseaux R, Di Carlo D. Elastomeric sensor surfaces for high-throughput single-cell force cytometry. Nat Biomed Eng 2018; 2:124-137. [PMID: 31015629 DOI: 10.1038/s41551-018-0193-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 01/09/2018] [Indexed: 11/09/2022]
Abstract
As cells with aberrant force-generating phenotypes can directly lead to disease, cellular force-generation mechanisms are high-value targets for new therapies. Here, we show that single-cell force sensors embedded in elastomers enable single-cell force measurements with ~100-fold improvement in throughput than was previously possible. The microtechnology is scalable and seamlessly integrates with the multi-well plate format, enabling highly parallelized time-course studies. In this regard, we show that airway smooth muscle cells isolated from fatally asthmatic patients have innately greater and faster force-generation capacity in response to stimulation than healthy control cells. By simultaneously tracing agonist-induced calcium flux and contractility in the same cell, we show that the calcium level is ultimately a poor quantitative predictor of cellular force generation. Finally, by quantifying phagocytic forces in thousands of individual human macrophages, we show that force initiation is a digital response (rather than a proportional one) to the proper immunogen. By combining mechanobiology at the single-cell level with high-throughput capabilities, this microtechnology can support drug-discovery efforts for clinical conditions associated with aberrant cellular force generation.
Collapse
Affiliation(s)
- Ivan Pushkarsky
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter Tseng
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Electrical Engineering and Computer Science, University of California, Los Angeles, Irvine, CA, USA
| | - Dylan Black
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bryan France
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lyndon Warfe
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - William F Jester
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Ryan K Trinh
- Department of Microbiology, Immunology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan Lin
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Philip O Scumpia
- Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sherie L Morrison
- Department of Microbiology, Immunology and Molecular Genetics and The Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Robert Damoiseaux
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Molecular and Medicinal Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA. .,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA. .,Department of Mechanical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Xiao K, Cao WB, Rong CH, Chen LG, Yang XX, Wen WJ, Qian PY, Hu ZL, Xu Y, Zhang Y. A novel assessment of the traction forces upon settlement of two typical marine fouling invertebrates using PDMS micropost arrays. Biol Open 2018; 7:bio030262. [PMID: 29242196 PMCID: PMC5829505 DOI: 10.1242/bio.030262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/28/2017] [Indexed: 11/30/2022] Open
Abstract
Marine biofouling poses a severe threat to maritime and aquaculture industries. To prevent the attachment of marine biofouling organisms on man-made structures, countless cost and effort was spent annually. In particular, most attention has been paid on the development of efficient and environmentally friendly fouling-resistant coatings, as well as larval settlement mechanism of several major biofouling invertebrates. In this study, polydimethylsiloxane (PDMS) micropost arrays were utilized as the settlement substrata and opposite tractions were identified during early settlement of the barnacle Amphibalanus amphitrite and the bryozoan Bugula neritina The settling A. amphitrite pushed the periphery microposts with an average traction force of 376.2 nN, while settling B. neritina pulled the periphery microposts with an average traction force of 205.9 nN. These micropost displacements are consistent with the body expansion of A. amphitrite during early post-settlement metamorphosis stage and elevation of wall epithelium of B. neritina during early pre-ancestrula stage, respectively. As such, the usage of micropost array may supplement the traditional histological approach to indicate the early settlement stages or even the initiation of larval settlement of marine fouling organisms, and could finally aid in the development of automatic monitoring platform for the real-time analysis on this complex biological process.
Collapse
Affiliation(s)
- Kang Xiao
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Engineering Research Center for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P.R. China
| | - Wen-Bin Cao
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, P.R. China
| | - Cu-Huang Rong
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Engineering Research Center for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
| | - Lian-Guo Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, P.R. China
| | - Xiao-Xue Yang
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, P.R. China
| | - Wei-Jia Wen
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, P.R. China
| | - Pei-Yuan Qian
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, P.R. China
| | - Zhang-Li Hu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Engineering Research Center for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Xu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Engineering Research Center for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
| | - Yu Zhang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Engineering Research Center for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, P.R. China
| |
Collapse
|