51
|
Cokić M, Bruegmann T, Sasse P, Malan D. Optogenetic Stimulation of G i Signaling Enables Instantaneous Modulation of Cardiomyocyte Pacemaking. Front Physiol 2022; 12:768495. [PMID: 34987414 PMCID: PMC8721037 DOI: 10.3389/fphys.2021.768495] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/18/2021] [Indexed: 01/01/2023] Open
Abstract
G-protein signaling pathways are central in the regulation of cardiac function in physiological and pathophysiological conditions. Their functional analysis through optogenetic techniques with selective expression of opsin proteins and activation by specific wavelengths allows high spatial and temporal precision. Here, we present the application of long wavelength-sensitive cone opsin (LWO) in cardiomyocytes for activation of the Gi signaling pathway by red light. Murine embryonic stem (ES) cells expressing LWO were generated and differentiated into beating cardiomyocytes in embryoid bodies (EBs). Illumination with red light (625 nm) led to an instantaneous decrease up to complete inhibition (84–99% effectivity) of spontaneous beating, but had no effect on control EBs. By using increasing light intensities with 10 s pulses, we determined a half maximal effective light intensity of 2.4 μW/mm2 and a maximum effect at 100 μW/mm2. Pre-incubation of LWO EBs with pertussis toxin completely inhibited the light effect proving the specificity for Gi signaling. Frequency reduction was mainly due to the activation of GIRK channels because the specific channel blocker tertiapin reduced the light effect by ~80%. Compared with pharmacological stimulation of M2 receptors with carbachol with slow kinetics (>30 s), illumination of LWO had an identical efficacy, but much faster kinetics (<1 s) in the activation and deactivation demonstrating the temporal advantage of optogenetic stimulation. Thus, LWO is an effective optogenetic tool for selective stimulation of the Gi signaling cascade in cardiomyocytes with red light, providing high temporal precision.
Collapse
Affiliation(s)
- Milan Cokić
- Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Tobias Bruegmann
- Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Philipp Sasse
- Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Malan
- Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| |
Collapse
|
52
|
Dokshokova L, Pianca N, Zaglia T, Mongillo M. Optogenetic Control of Heart Rhythm: Lightly Guiding the Cardiac Pace. Methods Mol Biol 2022; 2483:205-229. [PMID: 35286678 DOI: 10.1007/978-1-0716-2245-2_13] [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] [Indexed: 06/14/2023]
Abstract
It is well appreciated that, differently from skeletal muscles, the heart contracts independently from neurogenic inputs. However, the speed and force of heartbeats are finely modulated during stresses, emotions, and daily activities, by the autonomic neurons (both parasympathetic and sympathetic) which highly innervate the myocardium. Despite this aspect of cardiac physiology has been known for long, research has only recently shed light on the biophysical mechanisms underlying the meticulous adaptation of heart activity to the needs of the organism. A conceptual advancement in this regard has come from the use of optogenetics, a revolutionary methodology which allows to control the activity of a given excitable cell type, with high specificity, temporal and spatial resolution, within intact tissues and organisms. The method, widely affirmed in the field of neuroscience, has more recently been exploited also in research on heart physiology and pathology, including the study of the mechanisms regulating heart rhythm. The last point is the object of this book chapter which, starting from the description of the physiology of heart rhythm automaticity and the neurogenic modulation of heart rate, makes an excursus on the theoretical basis of such biotechnology (with its advantages and limitations), and presents a series of examples in cardiac and neuro-cardiac optogenetics.
Collapse
Affiliation(s)
- Lolita Dokshokova
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Nicola Pianca
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
53
|
Optogenetic manipulation of cardiac electrical dynamics using sub-threshold illumination: dissecting the role of cardiac alternans in terminating rapid rhythms. Basic Res Cardiol 2022; 117:25. [PMID: 35488105 PMCID: PMC9054908 DOI: 10.1007/s00395-022-00933-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/01/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023]
Abstract
Cardiac action potential (AP) shape and propagation are regulated by several key dynamic factors such as ion channel recovery and intracellular Ca2+ cycling. Experimental methods for manipulating AP electrical dynamics commonly use ion channel inhibitors that lack spatial and temporal specificity. In this work, we propose an approach based on optogenetics to manipulate cardiac electrical activity employing a light-modulated depolarizing current with intensities that are too low to elicit APs (sub-threshold illumination), but are sufficient to fine-tune AP electrical dynamics. We investigated the effects of sub-threshold illumination in isolated cardiomyocytes and whole hearts by using transgenic mice constitutively expressing a light-gated ion channel (channelrhodopsin-2, ChR2). We find that ChR2-mediated depolarizing current prolongs APs and reduces conduction velocity (CV) in a space-selective and reversible manner. Sub-threshold manipulation also affects the dynamics of cardiac electrical activity, increasing the magnitude of cardiac alternans. We used an optical system that uses real-time feedback control to generate re-entrant circuits with user-defined cycle lengths to explore the role of cardiac alternans in spontaneous termination of ventricular tachycardias (VTs). We demonstrate that VT stability significantly decreases during sub-threshold illumination primarily due to an increase in the amplitude of electrical oscillations, which implies that cardiac alternans may be beneficial in the context of self-termination of VT.
Collapse
|
54
|
Chen K, Ernst P, Liu XM, Zhou L. Optogenetic Studies of Mitochondria. Methods Mol Biol 2022; 2501:311-324. [PMID: 35857235 DOI: 10.1007/978-1-0716-2329-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
While optogenetic approaches have been widely used for remote control of cell membrane excitability and intracellular signaling pathways, their application in mitochondrial study has been limited, largely due to the challenge of effectively and specifically expressing heterologous light-gated rhodopsin channels in the mitochondria. Here, we describe the methods for expressing functional channelrhodopsin 2 (ChR2) proteins in the mitochondrial inner membrane with an unusually long mitochondrial leading sequence and characterizing optogenetic-mediated mitochondrial membrane potential (ΔΨm) depolarization. We then illustrate how this next-generation optogenetic approach can be used to study the effect of ΔΨm on mitochondrial functions such as mitophagy, programed cell death, and preconditioning-mediated cytoprotection. We anticipate that this innovative technology will enable new insights into the mechanisms by which changes in ΔΨm differentially impacts mitochondrial and cellular functions.
Collapse
Affiliation(s)
- Kai Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patrick Ernst
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaoguang Margaret Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lufang Zhou
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA.
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
55
|
Matsubara T, Yamashita T. Remote Optogenetics Using Up/Down-Conversion Phosphors. Front Mol Biosci 2021; 8:771717. [PMID: 34805279 PMCID: PMC8602066 DOI: 10.3389/fmolb.2021.771717] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
Microbial rhodopsins widely used for optogenetics are sensitive to light in the visible spectrum. As visible light is heavily scattered and absorbed by tissue, stimulating light for optogenetic control does not reach deep in the tissue irradiated from outside the subject body. Conventional optogenetics employs fiber optics inserted close to the target, which is highly invasive and poses various problems for researchers. Recent advances in material science integrated with neuroscience have enabled remote optogenetic control of neuronal activities in living animals using up- or down-conversion phosphors. The development of these methodologies has stimulated researchers to test novel strategies for less invasive, wireless control of cellular functions in the brain and other tissues. Here, we review recent reports related to these new technologies and discuss the current limitations and future perspectives toward the establishment of non-invasive optogenetics for clinical applications.
Collapse
Affiliation(s)
- Takanori Matsubara
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takayuki Yamashita
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
56
|
Müllenbroich MC, Kelly A, Acker C, Bub G, Bruegmann T, Di Bona A, Entcheva E, Ferrantini C, Kohl P, Lehnart SE, Mongillo M, Parmeggiani C, Richter C, Sasse P, Zaglia T, Sacconi L, Smith GL. Novel Optics-Based Approaches for Cardiac Electrophysiology: A Review. Front Physiol 2021; 12:769586. [PMID: 34867476 PMCID: PMC8637189 DOI: 10.3389/fphys.2021.769586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022] Open
Abstract
Optical techniques for recording and manipulating cellular electrophysiology have advanced rapidly in just a few decades. These developments allow for the analysis of cardiac cellular dynamics at multiple scales while largely overcoming the drawbacks associated with the use of electrodes. The recent advent of optogenetics opens up new possibilities for regional and tissue-level electrophysiological control and hold promise for future novel clinical applications. This article, which emerged from the international NOTICE workshop in 2018, reviews the state-of-the-art optical techniques used for cardiac electrophysiological research and the underlying biophysics. The design and performance of optical reporters and optogenetic actuators are reviewed along with limitations of current probes. The physics of light interaction with cardiac tissue is detailed and associated challenges with the use of optical sensors and actuators are presented. Case studies include the use of fluorescence recovery after photobleaching and super-resolution microscopy to explore the micro-structure of cardiac cells and a review of two photon and light sheet technologies applied to cardiac tissue. The emergence of cardiac optogenetics is reviewed and the current work exploring the potential clinical use of optogenetics is also described. Approaches which combine optogenetic manipulation and optical voltage measurement are discussed, in terms of platforms that allow real-time manipulation of whole heart electrophysiology in open and closed-loop systems to study optimal ways to terminate spiral arrhythmias. The design and operation of optics-based approaches that allow high-throughput cardiac electrophysiological assays is presented. Finally, emerging techniques of photo-acoustic imaging and stress sensors are described along with strategies for future development and establishment of these techniques in mainstream electrophysiological research.
Collapse
Affiliation(s)
| | - Allen Kelly
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Corey Acker
- Center for Cell Analysis and Modeling, UConn Health, Farmington, CT, United States
| | - Gil Bub
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Tobias Bruegmann
- Institute for Cardiovascular Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | | | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Stephan E. Lehnart
- Heart Research Center Göttingen, University Medical Center Göttingen, Göttingen, Germany
- Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | | | - Claudia Richter
- German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Leonardo Sacconi
- European Laboratory for Nonlinear Spectroscopy, Sesto Fiorentino, Italy
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
- National Institute of Optics, National Research Council, Florence, Italy
| | - Godfrey L. Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
57
|
Kim WS, Hong S, Park SI. Robust, wireless gastric optogenetic implants for the study of peripheral pathways and applications in obesity . ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:5742-5746. [PMID: 34892424 DOI: 10.1109/embc46164.2021.9629753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Optogenetics has the potential to transform the study of organ functions in the peripheral nervous system via relatively easy access to the nerves and a direct link between the brain and organ systems. Implementation typically requires a static skeletal feature for the securement of a fiber. Unfortunately, the soft nature of peripheral nervous systems makes the wired fiber-optic approach less ideal for the study of the peripheral nervous system. Existing wireless approaches could bypass some constraints associated with optical fibers and thereby offer organ specificity. However, they suffer from durability loss due to considerable biological strains and unable to perform longitudinal experiments. Here, we propose a new class of wireless gastric optogenetic implant for identifying signaling pathways, in particular viscerosensory pathways, that can regulate food intake to treat obesity. Robust, wireless gastric optogenetic implants with a tubing-assisted U-shaped tether directly interface with nerve endings in the stomach with chronic stability in operation (> 100 kilocycles) and allows for optogenetic stimulations of vagus nerves in a freely behaving animal. We demonstrated utilities of the proposed wireless device in in vivo experiments. Results suggest the potential for identifying interventions for the treatment of obesity.Clinical Relevance - Identification of the roles of subpopulations in viscerosensory pathways would provide the platform for the development of better therapeutics for the treatment of obesity.
Collapse
|
58
|
Lin Y, Yao Y, Zhang W, Fang Q, Zhang L, Zhang Y, Xu Y. Applications of upconversion nanoparticles in cellular optogenetics. Acta Biomater 2021; 135:1-12. [PMID: 34461347 DOI: 10.1016/j.actbio.2021.08.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 08/07/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022]
Abstract
Upconversion-mediated optogenetics is an emerging powerful technique to remotely control and manipulate the deep-tissue protein functions and signaling pathway activation. This technique uses lanthanide upconversion nanoparticles (UCNPs) as light transducers and through near-infrared light to indirectly activate the traditional optogenetic proteins. With the merits of high spatiotemporal resolution and minimal invasiveness, this technique enables cell-type specific manipulation of cellular activities in deep tissues as well as in living animals. In this review, we introduce the latest development of optogenetic modules and UCNPs, with emphasis on the integration of UCNPs with cellular optogenetics and their biomedical applications on the control of neural/brain activity, cancer therapy and cardiac optogenetics in vivo. Furthermore, we analyze the current developed strategies to optimize and advance the upconversion-mediated optogenetics and discuss the remaining challenges of its further applications in biomedical study and clinical translational research. STATEMENT OF SIGNIFICANCE: Optogenetics harnesses photoactivatable proteins to optically stimulate and control intracellular activities. UCNPs-mediated NIR-activatable optogenetics uses lanthanide upconversion nanoparticles (UCNPs) as light transducers and utilizes near-infrared (NIR) light to indirectly activate the traditional optogenetic proteins. The integration of UCNPs with cellular optogenetics has showed great promise in biomedical applications in regulating neural/brain activity, cancer therapy and cardiac optogenetics in vivo. The evolution and optimization of functional UCNPs and the discovery and engineering of novel optogenetic modules would both contribute to the advance of such unique hybrid technology, which may lead to discoveries in biomedical research and provide new treatments for human diseases.
Collapse
Affiliation(s)
- Yinyan Lin
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yuanfa Yao
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Wanmei Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qiuyu Fang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Luhao Zhang
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Yan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China; Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| |
Collapse
|
59
|
Madrid MK, Brennan JA, Yin RT, Knight HS, Efimov IR. Advances in Implantable Optogenetic Technology for Cardiovascular Research and Medicine. Front Physiol 2021; 12:720190. [PMID: 34675815 PMCID: PMC8523791 DOI: 10.3389/fphys.2021.720190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Optogenetic technology provides researchers with spatiotemporally precise tools for stimulation, sensing, and analysis of function in cells, tissues, and organs. These tools can offer low-energy and localized approaches due to the use of the transgenically expressed light gated cation channel Channelrhodopsin-2 (ChR2). While the field began with many neurobiological accomplishments it has also evolved exceptionally well in animal cardiac research, both in vitro and in vivo. Implantable optical devices are being extensively developed to study particular electrophysiological phenomena with the precise control that optogenetics provides. In this review, we highlight recent advances in novel implantable optogenetic devices and their feasibility in cardiac research. Furthermore, we also emphasize the difficulties in translating this technology toward clinical applications and discuss potential solutions for successful clinical translation.
Collapse
Affiliation(s)
- Micah K Madrid
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Jaclyn A Brennan
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Rose T Yin
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Helen S Knight
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| |
Collapse
|
60
|
Rieger M, Dellenbach C, Vom Berg J, Beil-Wagner J, Maguy A, Rohr S. Enabling comprehensive optogenetic studies of mouse hearts by simultaneous opto-electrical panoramic mapping and stimulation. Nat Commun 2021; 12:5804. [PMID: 34608155 PMCID: PMC8490461 DOI: 10.1038/s41467-021-26039-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/15/2021] [Indexed: 11/09/2022] Open
Abstract
During the last decade, cardiac optogenetics has turned into an essential tool for investigating cardiac function in general and for assessing functional interactions between different myocardial cell types in particular. To advance exploitation of the unique research opportunities offered by this method, we develop a panoramic opto-electrical measurement and stimulation (POEMS) system for mouse hearts. The core of the experimental platform is composed of 294 optical fibers and 64 electrodes that form a cup which embraces the entire ventricular surface of mouse hearts and enables straightforward ‘drop&go’ experimentation. The flexible assignment of fibers and electrodes to recording or stimulation tasks permits a precise tailoring of experiments to the specific requirements of individual optogenetic constructs thereby avoiding spectral congestion. Validation experiments with hearts from transgenic animals expressing the optogenetic voltage reporters ASAP1 and ArcLight-Q239 demonstrate concordance of simultaneously recorded panoramic optical and electrical activation maps. The feasibility of single fiber optical stimulation is proven with hearts expressing the optogenetic voltage actuator ReaChR. Adaptation of the POEMS system to larger hearts and incorporation of additional sensors can be achieved by redesigning the system-core accordingly. Current cardiac mapping systems provide either electrical or optical readouts. Here the authors report a panoramic opto-electrical measurement and stimulation (POEMS) system which embraces the entire ventricular surface of mouse hearts, allowing flexible combinations of optical and electrical recording and stimulation modalities.
Collapse
Affiliation(s)
- Michael Rieger
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, Switzerland
| | | | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Jane Beil-Wagner
- Institute of Laboratory Animal Science, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| | - Ange Maguy
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, Switzerland
| | - Stephan Rohr
- Department of Physiology, University of Bern, Bühlplatz 5, Bern, Switzerland.
| |
Collapse
|
61
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
62
|
Zhan H, Wang Z, Lin J, Yu Y, Xia L. Optogenetic actuation in ChR2-transduced fibroblasts alter excitation-contraction coupling and mechano-electric feedback in coupled cardiomyocytes: a computational modeling study. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:8354-8373. [PMID: 34814303 DOI: 10.3934/mbe.2021414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With the help of the conventional electrical method and the growing optogenetic technology, cardiac fibroblasts (Fbs) have been verified to couple electrically with working myocytes and bring electrophysiological remodeling changes in them. The intrinsic properties of cardiac functional autoregulation represented by excitation-contraction coupling (ECC) and mechano-electric feedback (MEF) have also been extensively studied. However, the roles of optogenetic stimulation on the characteristics of ECC and MEF in cardiomyocytes (CMs) coupled with Fbs have been barely investigated. In this study, we proposed a combined model composed of three modules to explore these influences. Simulation results showed that (1) during ECC, an increased light duration (LD) strengthened the inflow of ChR2 current and prolonged action potential duration (APD), and extended durations of twitch and internal sarcomere deformation through the decreased dissociation of calcium with troponin C (CaTnC) complexes and the prolonged duration of Xb attachment-detachment; (2) during MEF, an increased LD was followed by a longer muscle twitch and deformation, and led to APD prolongation through the inward ChR2 current and its inward rectification kinetics, which far outweighed the effects of the delaying dissociation of CaTnC complexes and the prolonged reverse mode of Na+-Ca2+ exchange on AP shortening; (3) due to the ChR2 current's rectification feature, enhancing the light irradiance (LI) brought slight variations in peak or valley values of electrophysiological and mechanical parameters while did not change durations of AP and twitch and muscle deformation in both ECC and MEF. In conclusion, the inward ChR2 current and its inward rectification feature were found to affect significantly the durations of AP and twitch in both ECC and MEF. The roles of optogenetic actuation on both ECC and MEF should be considered in future cardiac computational optogenetics at the tissue and organ scale.
Collapse
Affiliation(s)
- Heqing Zhan
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Zefeng Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jialun Lin
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Yuanbo Yu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, China
| | - Ling Xia
- Key Laboratory for Biomedical Engineering of Ministry of Education, Institute of Biomedical Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
63
|
Credi C, Balducci V, Munagala U, Cianca C, Bigiarini S, de Vries AAF, Loew LM, Pavone FS, Cerbai E, Sartiani L, Sacconi L. Fast Optical Investigation of Cardiac Electrophysiology by Parallel Detection in Multiwell Plates. Front Physiol 2021; 12:692496. [PMID: 34539428 PMCID: PMC8446431 DOI: 10.3389/fphys.2021.692496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
Current techniques for fast characterization of cardiac electrophysiology employ optical technologies to control and monitor action potential features of single cells or cellular monolayers placed in multiwell plates. High-speed investigation capacities are commonly achieved by serially analyzing well after well employing fully automated fluorescence microscopes. Here, we describe an alternative cost-effective optical approach (MULTIPLE) that exploits high-power LED arrays to globally illuminate a culture plate and an sCMOS sensor for parallel detection of the fluorescence coming from multiple wells. MULTIPLE combines optical detection of action potentials using a red-shifted voltage-sensitive fluorescent dye (di-4-ANBDQPQ) with optical stimulation, employing optogenetic actuators, to ensure excitation of cardiomyocytes at constant rates. MULTIPLE was first characterized in terms of interwell uniformity of the illumination intensity and optical detection performance. Then, it was applied for probing action potential features in HL-1 cells (i.e., mouse atrial myocyte-like cells) stably expressing the blue light-activatable cation channel CheRiff. Under proper stimulation conditions, we were able to accurately measure action potential dynamics across a 24-well plate with variability across the whole plate of the order of 10%. The capability of MULTIPLE to detect action potential changes across a 24-well plate was demonstrated employing the selective K v 11.1 channel blocker (E-4031), in a dose titration experiment. Finally, action potential recordings were performed in spontaneous beating human induced pluripotent stem cell derived cardiomyocytes following pharmacological manipulation of their beating frequency. We believe that the simplicity of the presented optical scheme represents a valid complement to sophisticated and expensive state-of-the-art optical systems for high-throughput cardiac electrophysiological investigations.
Collapse
Affiliation(s)
- Caterina Credi
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, Florence, Italy
| | - Valentina Balducci
- Department of Neurosciences, Psychology, Drugs and Child Health, University of Florence, Florence, Italy
| | - U Munagala
- Department of Neurosciences, Psychology, Drugs and Child Health, University of Florence, Florence, Italy.,Core Research Laboratory, ISPRO, Florence, Italy
| | - C Cianca
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy
| | - S Bigiarini
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Leslie M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Francesco S Pavone
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, Florence, Italy.,Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy
| | - Elisabetta Cerbai
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy.,Department of Neurosciences, Psychology, Drugs and Child Health, University of Florence, Florence, Italy
| | - Laura Sartiani
- Department of Neurosciences, Psychology, Drugs and Child Health, University of Florence, Florence, Italy
| | - Leonardo Sacconi
- European Laboratory for Non-linear Spectroscopy, Sesto Fiorentino, Italy.,National Institute of Optics, National Research Council, Florence, Italy
| |
Collapse
|
64
|
Ördög B, Teplenin A, De Coster T, Bart CI, Dekker SO, Zhang J, Ypey DL, de Vries AAF, Pijnappels DA. The Effects of Repetitive Use and Pathological Remodeling on Channelrhodopsin Function in Cardiomyocytes. Front Physiol 2021; 12:710020. [PMID: 34539432 PMCID: PMC8448166 DOI: 10.3389/fphys.2021.710020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/23/2021] [Indexed: 11/13/2022] Open
Abstract
Aim: Channelrhodopsins (ChRs) are a large family of light-gated ion channels with distinct properties, which is of great importance in the selection of a ChR variant for a given application. However, data to guide such selection for cardiac optogenetic applications are lacking. Therefore, we investigated the functioning of different ChR variants in normal and pathological hypertrophic cardiomyocytes subjected to various illumination protocols. Methods and Results: Isolated neonatal rat ventricular cardiomyocytes (NRVMs) were transduced with lentiviral vectors to express one of the following ChR variants: H134R, CatCh, ReaChR, or GtACR1. NRVMs were treated with phenylephrine (PE) to induce pathological hypertrophy (PE group) or left untreated [control (CTL) group]. In these groups, ChR currents displayed unique and significantly different properties for each ChR variant on activation by a single 1-s light pulse (1 mW/mm2: 470, 565, or 617 nm). The concomitant membrane potential (Vm) responses also showed a ChR variant-specific profile, with GtACR1 causing a slight increase in average Vm during illumination (Vplateau: −38 mV) as compared with a Vplateau > −20 mV for the other ChR variants. On repetitive activation at increasing frequencies (10-ms pulses at 1–10 Hz for 30 s), peak currents, which are important for cardiac pacing, decreased with increasing activation frequencies by 17–78% (p < 0.05), while plateau currents, which are critical for arrhythmia termination, decreased by 10–75% (p < 0.05), both in a variant-specific manner. In contrast, the corresponding Vplateau remained largely stable. Importantly, current properties and Vm responses were not statistically different between the PE and CTL groups, irrespective of the variant used (p > 0.05). Conclusion: Our data show that ChR variants function equally well in cell culture models of healthy and pathologically hypertrophic myocardium but show strong, variant-specific use-dependence. This use-dependent nature of ChR function should be taken into account during the design of cardiac optogenetic studies and the interpretation of the experimental findings thereof.
Collapse
Affiliation(s)
- Balázs Ördög
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Alexander Teplenin
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Tim De Coster
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Cindy I Bart
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Sven O Dekker
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Juan Zhang
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Dirk L Ypey
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Antoine A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
65
|
Ochs AR, Karathanos TV, Trayanova NA, Boyle PM. Optogenetic Stimulation Using Anion Channelrhodopsin (GtACR1) Facilitates Termination of Reentrant Arrhythmias With Low Light Energy Requirements: A Computational Study. Front Physiol 2021; 12:718622. [PMID: 34526912 PMCID: PMC8435849 DOI: 10.3389/fphys.2021.718622] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
Optogenetic defibrillation of hearts expressing light-sensitive cation channels (e.g., ChR2) has been proposed as an alternative to conventional electrotherapy. Past modeling work has shown that ChR2 stimulation can depolarize enough myocardium to interrupt arrhythmia, but its efficacy is limited by light attenuation and high energy needs. These shortcomings may be mitigated by using new optogenetic proteins like Guillardia theta Anion Channelrhodopsin (GtACR1), which produces a repolarizing outward current upon illumination. Accordingly, we designed a study to assess the feasibility of GtACR1-based optogenetic arrhythmia termination in human hearts. We conducted electrophysiological simulations in MRI-based atrial or ventricular models (n = 3 each), with pathological remodeling from atrial fibrillation or ischemic cardiomyopathy, respectively. We simulated light sensitization via viral gene delivery of three different opsins (ChR2, red-shifted ChR2, GtACR1) and uniform endocardial illumination at the appropriate wavelengths (blue, red, or green light, respectively). To analyze consistency of arrhythmia termination, we varied pulse timing (three evenly spaced intervals spanning the reentrant cycle) and intensity (atrial: 0.001–1 mW/mm2; ventricular: 0.001–10 mW/mm2). In atrial models, GtACR1 stimulation with 0.005 mW/mm2 green light consistently terminated reentry; this was 10–100x weaker than the threshold levels for ChR2-mediated defibrillation. In ventricular models, defibrillation was observed in 2/3 models for GtACR1 stimulation at 0.005 mW/mm2 (100–200x weaker than ChR2 cases). In the third ventricular model, defibrillation failed in nearly all cases, suggesting that attenuation issues and patient-specific organ/scar geometry may thwart termination in some cases. Across all models, the mechanism of GtACR1-mediated defibrillation was voltage forcing of illuminated tissue toward the modeled channel reversal potential of −40 mV, which made propagation through affected regions impossible. Thus, our findings suggest GtACR1-based optogenetic defibrillation of the human heart may be feasible with ≈2–3 orders of magnitude less energy than ChR2.
Collapse
Affiliation(s)
- Alexander R Ochs
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Thomas V Karathanos
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, United States
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, United States
| |
Collapse
|
66
|
Scalco A, Moro N, Mongillo M, Zaglia T. Neurohumoral Cardiac Regulation: Optogenetics Gets Into the Groove. Front Physiol 2021; 12:726895. [PMID: 34531763 PMCID: PMC8438220 DOI: 10.3389/fphys.2021.726895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022] Open
Abstract
The cardiac autonomic nervous system (ANS) is the main modulator of heart function, adapting contraction force, and rate to the continuous variations of intrinsic and extrinsic environmental conditions. While the parasympathetic branch dominates during rest-and-digest sympathetic neuron (SN) activation ensures the rapid, efficient, and repeatable increase of heart performance, e.g., during the "fight-or-flight response." Although the key role of the nervous system in cardiac homeostasis was evident to the eyes of physiologists and cardiologists, the degree of cardiac innervation, and the complexity of its circuits has remained underestimated for too long. In addition, the mechanisms allowing elevated efficiency and precision of neurogenic control of heart function have somehow lingered in the dark. This can be ascribed to the absence of methods adequate to study complex cardiac electric circuits in the unceasingly moving heart. An increasing number of studies adds to the scenario the evidence of an intracardiac neuron system, which, together with the autonomic components, define a little brain inside the heart, in fervent dialogue with the central nervous system (CNS). The advent of optogenetics, allowing control the activity of excitable cells with cell specificity, spatial selectivity, and temporal resolution, has allowed to shed light on basic neuro-cardiology. This review describes how optogenetics, which has extensively been used to interrogate the circuits of the CNS, has been applied to untangle the knots of heart innervation, unveiling the cellular mechanisms of neurogenic control of heart function, in physiology and pathology, as well as those participating to brain-heart communication, back and forth. We discuss existing literature, providing a comprehensive view of the advancement in the understanding of the mechanisms of neurogenic heart control. In addition, we weigh the limits and potential of optogenetics in basic and applied research in neuro-cardiology.
Collapse
Affiliation(s)
- Arianna Scalco
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Nicola Moro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Tania Zaglia
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
67
|
Keshmiri Neghab H, Soheilifar MH, Grusch M, Ortega MM, Esmaeeli Djavid G, Saboury AA, Goliaei B. The state of the art of biomedical applications of optogenetics. Lasers Surg Med 2021; 54:202-216. [PMID: 34363230 DOI: 10.1002/lsm.23463] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Optogenetics has opened new insights into biomedical research with the ability to manipulate and control cellular activity using light in combination with genetically engineered photosensitive proteins. By stimulating with light, this method provides high spatiotemporal and high specificity resolution, which is in contrast to conventional pharmacological or electrical stimulation. Optogenetics was initially introduced to control neural activities but was gradually extended to other biomedical fields. STUDY DESIGN In this paper, firstly, we summarize the current optogenetic tools stimulated by different light sources, including lasers, light-emitting diodes, and laser diodes. Second, we outline the variety of biomedical applications of optogenetics not only for neuronal circuits but also for various kinds of cells and tissues from cardiomyocytes to ganglion cells. Furthermore, we highlight the potential of this technique for treating neurological disorders, cardiac arrhythmia, visual impairment, hearing loss, and urinary bladder diseases as well as clarify the mechanisms underlying cancer progression and control of stem cell differentiation. CONCLUSION We sought to summarize the various types of promising applications of optogenetics to treat a broad spectrum of disorders. It is conceivable to expect that optogenetics profits a growing number of patients suffering from a range of different diseases in the near future.
Collapse
Affiliation(s)
- Hoda Keshmiri Neghab
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | | | - Michael Grusch
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Manoela Marques Ortega
- Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, São Francisco University, Bragança Paulista, São Paulo, Brazil
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Ali Akbar Saboury
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Bahram Goliaei
- Department of Biophysics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
68
|
Abstract
Conduction disorders and arrhythmias remain difficult to treat and are increasingly prevalent owing to the increasing age and body mass of the general population, because both are risk factors for arrhythmia. Many of the underlying conditions that give rise to arrhythmia - including atrial fibrillation and ventricular arrhythmia, which frequently occur in patients with acute myocardial ischaemia or heart failure - can have an inflammatory component. In the past, inflammation was viewed mostly as an epiphenomenon associated with arrhythmia; however, the recently discovered inflammatory and non-canonical functions of cardiac immune cells indicate that leukocytes can be arrhythmogenic either by altering tissue composition or by interacting with cardiomyocytes; for example, by changing their phenotype or perhaps even by directly interfering with conduction. In this Review, we discuss the electrophysiological properties of leukocytes and how these cells relate to conduction in the heart. Given the thematic parallels, we also summarize the interactions between immune cells and neural systems that influence information transfer, extrapolating findings from the field of neuroscience to the heart and defining common themes. We aim to bridge the knowledge gap between electrophysiology and immunology, to promote conceptual connections between these two fields and to explore promising opportunities for future research.
Collapse
|
69
|
Odening KE, Gomez AM, Dobrev D, Fabritz L, Heinzel FR, Mangoni ME, Molina CE, Sacconi L, Smith G, Stengl M, Thomas D, Zaza A, Remme CA, Heijman J. ESC working group on cardiac cellular electrophysiology position paper: relevance, opportunities, and limitations of experimental models for cardiac electrophysiology research. Europace 2021; 23:1795-1814. [PMID: 34313298 DOI: 10.1093/europace/euab142] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are a major cause of death and disability. A large number of experimental cell and animal models have been developed to study arrhythmogenic diseases. These models have provided important insights into the underlying arrhythmia mechanisms and translational options for their therapeutic management. This position paper from the ESC Working Group on Cardiac Cellular Electrophysiology provides an overview of (i) currently available in vitro, ex vivo, and in vivo electrophysiological research methodologies, (ii) the most commonly used experimental (cellular and animal) models for cardiac arrhythmias including relevant species differences, (iii) the use of human cardiac tissue, induced pluripotent stem cell (hiPSC)-derived and in silico models to study cardiac arrhythmias, and (iv) the availability, relevance, limitations, and opportunities of these cellular and animal models to recapitulate specific acquired and inherited arrhythmogenic diseases, including atrial fibrillation, heart failure, cardiomyopathy, myocarditis, sinus node, and conduction disorders and channelopathies. By promoting a better understanding of these models and their limitations, this position paper aims to improve the quality of basic research in cardiac electrophysiology, with the ultimate goal to facilitate the clinical translation and application of basic electrophysiological research findings on arrhythmia mechanisms and therapies.
Collapse
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland.,Institute of Physiology, University of Bern, Bern, Switzerland
| | - Ana-Maria Gomez
- Signaling and cardiovascular pathophysiology-UMR-S 1180, Inserm, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.,Department of Cardiology, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Leonardo Sacconi
- National Institute of Optics and European Laboratory for Non Linear Spectroscopy, Italy.,Institute for Experimental Cardiovascular Medicine, University Freiburg, Germany
| | - Godfrey Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Milan Stengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Dierk Thomas
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Center for Heart Rhythm Disorders (HCR), University Hospital Heidelberg, Heidelberg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Antonio Zaza
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
70
|
Majumder R, Mohamed Nazer AN, Panfilov AV, Bodenschatz E, Wang Y. Electrophysiological Characterization of Human Atria: The Understated Role of Temperature. Front Physiol 2021; 12:639149. [PMID: 34366877 PMCID: PMC8346027 DOI: 10.3389/fphys.2021.639149] [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: 12/08/2020] [Accepted: 04/01/2021] [Indexed: 11/13/2022] Open
Abstract
Ambient temperature has a profound influence on cellular electrophysiology through direct control over the gating mechanisms of different ion channels. In the heart, low temperature is known to favor prolongation of the action potential. However, not much is known about the influence of temperature on other important characterization parameters such as the resting membrane potential (RMP), excitability, morphology and characteristics of the action potential (AP), restitution properties, conduction velocity (CV) of signal propagation, etc. Here we present the first, detailed, systematic in silico study of the electrophysiological characterization of cardiomyocytes from different regions of the normal human atria, based on the effects of ambient temperature (5-50°C). We observe that RMP decreases with increasing temperature. At ~ 48°C, the cells lose their excitability. Our studies show that different parts of the atria react differently to the same changes in temperature. In tissue simulations a drop in temperature correlated positively with a decrease in CV, but the decrease was region-dependent, as expected. In this article we show how this heterogeneous response can provide an explanation for the development of a proarrhythmic substrate during mild hypothermia. We use the above concept to propose a treatment strategy for atrial fibrillation that involves severe hypothermia in specific regions of the heart for a duration of only ~ 200 ms.
Collapse
Affiliation(s)
- Rupamanjari Majumder
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | | | - Alexander V Panfilov
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia.,Department of Physics and Astronomy, Ghent University, Ghent, Belgium.,Laboratory of Computational Biology and Medicine, Ural Federal University, Yekaterinburg, Russia
| | - Eberhard Bodenschatz
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Laboratory of Atomic and Solid-State Physics and Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, United States
| | - Yong Wang
- Laboratory for Fluid Physics, Pattern Formation and Biocomplexity, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
71
|
Wichert N, Witt M, Blume C, Scheper T. Clinical applicability of optogenetic gene regulation. Biotechnol Bioeng 2021; 118:4168-4185. [PMID: 34287844 DOI: 10.1002/bit.27895] [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: 11/24/2020] [Revised: 05/27/2021] [Accepted: 07/13/2021] [Indexed: 11/10/2022]
Abstract
The field of optogenetics is rapidly growing in relevance and number of developed tools. Among other things, the optogenetic repertoire includes light-responsive ion channels and methods for gene regulation. This review will be confined to the optogenetic control of gene expression in mammalian cells as suitable models for clinical applications. Here optogenetic gene regulation might offer an excellent method for spatially and timely regulated gene and protein expression in cell therapeutic approaches. Well-known systems for gene regulation, such as the LOV-, CRY2/CIB-, PhyB/PIF-systems, as well as other, in mammalian cells not yet fully established systems, will be described. Advantages and disadvantages with regard to clinical applications are outlined in detail. Among the many unanswered questions concerning the application of optogenetics, we discuss items such as the use of exogenous chromophores and their effects on the biology of the cells and methods for a gentle, but effective gene transfection method for optogenetic tools for in vivo applications.
Collapse
Affiliation(s)
- Nina Wichert
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Martin Witt
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Cornelia Blume
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| | - Thomas Scheper
- Insitute of Technical Chemistry, Leibniz University of Hannover, Hannover, Germany
| |
Collapse
|
72
|
Percival SJ, Russo S, Priest C, Hill RC, Ohlhausen JA, Small LJ, Rempe SB, Spoerke ED. Bio-inspired incorporation of phenylalanine enhances ionic selectivity in layer-by-layer deposited polyelectrolyte films. SOFT MATTER 2021; 17:6315-6325. [PMID: 33982047 DOI: 10.1039/d1sm00134e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The addition of a common amino acid, phenylalanine, to a Layer-by-Layer (LbL) deposited polyelectrolyte (PE) film on a nanoporous membrane can increase its ionic selectivity over a PE film without the added amino acid. The addition of phenylalanine is inspired by detailed knowledge of the structure of the channelrhodopsins family of protein ion channels, where phenylalanine plays an instrumental role in facilitating sodium ion transport. The normally deposited and crosslinked PE films increase the cationic selectivity of a support membrane in a controllable manner where higher selectivity is achieved with thicker PE coatings, which in turn also increases the ionic resistance of the membrane. The increased ionic selectivity is desired while the increased resistance is not. We show that through incorporation of phenylalanine during the LbL deposition process, in solutions of NaCl with concentrations ranging from 0.1 to 100 mM, the ionic selectivity can be increased independently of the membrane resistance. Specifically, the addition is shown to increase the cationic transference of the PE films from 81.4% to 86.4%, an increase on par with PE films that are nearly triple the thickness while exhibiting much lower resistance compared to the thicker coatings, where the phenylalanine incorporated PE films display an area specific resistance of 1.81 Ω cm2 in 100 mM NaCl while much thicker PE membranes show a higher resistance of 2.75 Ω cm2 in the same 100 mM NaCl solution.
Collapse
Affiliation(s)
- Stephen J Percival
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Sara Russo
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Chad Priest
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Ryan C Hill
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - James A Ohlhausen
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Leo J Small
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Susan B Rempe
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| | - Erik D Spoerke
- Sandia National Laboratories, PO Box 5800, MS 1411, Albuquerque, NM 87185, USA.
| |
Collapse
|
73
|
Dong X, Kheiri S, Lu Y, Xu Z, Zhen M, Liu X. Toward a living soft microrobot through optogenetic locomotion control of Caenorhabditis elegans. Sci Robot 2021; 6:6/55/eabe3950. [PMID: 34193562 DOI: 10.1126/scirobotics.abe3950] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
Learning from the locomotion of natural organisms is one of the most effective strategies for designing microrobots. However, the development of bioinspired microrobots is still challenging because of technical bottlenecks such as design and seamless integration of high-performance actuation mechanism and high-density energy source for untethered locomotion. Directly harnessing the activation energy and intelligence of living tissues in synthetic micromachines provides an alternative route to developing biohybrid microrobots. Here, we propose an approach to engineering the genetic and nervous systems of a nematode worm, Caenorhabditis elegans, and creating an untethered, highly controllable living soft microrobot (called "RoboWorm"). A living worm is engineered through optogenetic and biochemical methods to shut down the signal transmissions between its neuronal and muscular systems while its muscle cells still remain optically excitable. Through dynamic modeling and experimental verification of the worm crawling, we found that the phase difference between the worm body curvature and the muscular activation pattern generates the thrust force for crawling locomotion. By reproducing the phase difference via optogenetic excitation of the worm body muscles, we emulated the major worm crawling behaviors in a controllable manner. Furthermore, with real-time visual feedback of the worm crawling, we realized closed-loop regulation of the movement direction and destination of single worms. This technology may facilitate scientific studies on the biophysics and neural basis of crawling locomotion of C. elegans and other nematode species.
Collapse
Affiliation(s)
- Xianke Dong
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario M5S 3G8, Canada.,Department of Mechanical Engineering, McGill University, 817 Sherbrooke Street West, Montreal, Quebec H3A 2K6, Canada
| | - Sina Kheiri
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario M5S 3G8, Canada
| | - Yangning Lu
- Lunenfeld-Tanenbaum Research Institute, 600 University Avenue, Room 870, Toronto, Ontario M5G 1X5, Canada.,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Zhaoyi Xu
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario M5S 3G8, Canada
| | - Mei Zhen
- Lunenfeld-Tanenbaum Research Institute, 600 University Avenue, Room 870, Toronto, Ontario M5G 1X5, Canada.,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Xinyu Liu
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, Ontario M5S 3G8, Canada. .,Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
74
|
Funken M, Bruegmann T, Sasse P. Selective optogenetic stimulation of fibroblasts enables quantification of hetero-cellular coupling to cardiomyocytes in a three-dimensional model of heart tissue. Europace 2021; 22:1590-1599. [PMID: 32808019 DOI: 10.1093/europace/euaa128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/27/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Besides providing mechanical stability, fibroblasts in the heart could modulate the electrical properties of cardiomyocytes. Here, we aim to develop a three-dimensional hetero-cellular model to analyse the electric interaction between fibroblasts and human cardiomyocytes in vitro using selective optogenetic de- or hyperpolarization of fibroblasts. METHODS AND RESULTS NIH3T3 cell lines expressing the light-sensitive ion channel Channelrhodopsin2 or the light-induced proton pump Archaerhodopsin were generated for optogenetic depolarization or hyperpolarization, respectively, and characterized by patch clamp. Cardiac bodies consisting of 50% fibroblasts and 50% human pluripotent stem cell-derived cardiomyocytes were analysed by video microscopy and membrane potential was measured with sharp electrodes. Myofibroblast activation in cardiac bodies was enhanced by transforming growth factor-β1 (TGF-β1)-stimulation. Connexin-43 expression was analysed by qPCR and fluorescence recovery after photobleaching. Illumination of Channelrhodopsin2 or Archaerhodopsin expressing fibroblasts induced inward currents and depolarization or outward currents and hyperpolarization. Transforming growth factor-β1-stimulation elevated connexin-43 expression and increased cell-cell coupling between fibroblasts as well as increased basal beating frequency and cardiomyocyte resting membrane potential in cardiac bodies. Illumination of cardiac bodies generated with Channelrhodopsin2 fibroblasts accelerated spontaneous beating, especially after TGF-β1-stimulation. Illumination of cardiac bodies prepared with Archaerhodopsin expressing fibroblasts led to hyperpolarization of cardiomyocytes and complete block of spontaneous beating after TGF-β1-stimulation. Effects of light were significantly smaller without TGF-β1-stimulation. CONCLUSION Transforming growth factor-β1-stimulation leads to increased hetero-cellular coupling and optogenetic hyperpolarization of fibroblasts reduces TGF-β1 induced effects on cardiomyocyte spontaneous activity. Optogenetic membrane potential manipulation selectively in fibroblasts in a new hetero-cellular cardiac body model allows direct quantification of fibroblast-cardiomyocyte coupling in vitro.
Collapse
Affiliation(s)
- Maximilian Funken
- Institute of Physiology I, Medical Faculty, University of Bonn, Nussallee 11, 53115 Bonn, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, Nussallee 11, 53115 Bonn, Germany.,Institute for Cardiovascular Physiology, University Medical Center Goettingen, Goettingen, Germany.,DZHK e. V. (German Center for Cardiovascular Research), Partner Site Goettingen, Goettingen, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Nussallee 11, 53115 Bonn, Germany
| |
Collapse
|
75
|
Chua CJ, Han JL, Li W, Liu W, Entcheva E. Integration of Engineered "Spark-Cell" Spheroids for Optical Pacing of Cardiac Tissue. Front Bioeng Biotechnol 2021; 9:658594. [PMID: 34222210 PMCID: PMC8249938 DOI: 10.3389/fbioe.2021.658594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/18/2021] [Indexed: 12/03/2022] Open
Abstract
Optogenetic methods for pacing of cardiac tissue can be realized by direct genetic modification of the cardiomyocytes to express light-sensitive actuators, such as channelrhodopsin-2, ChR2, or by introduction of light-sensitized non-myocytes that couple to the cardiac cells and yield responsiveness to optical pacing. In this study, we engineer three-dimensional “spark cells” spheroids, composed of ChR2-expressing human embryonic kidney cells (from 100 to 100,000 cells per spheroid), and characterize their morphology as function of cell density and time. These “spark-cell” spheroids are then deployed to demonstrate site-specific optical pacing of human stem-cell-derived cardiomyocytes (hiPSC-CMs) in 96-well format using non-localized light application and all-optical electrophysiology with voltage and calcium small-molecule dyes or genetically encoded sensors. We show that the spheroids can be handled using liquid pipetting and can confer optical responsiveness of cardiac tissue earlier than direct viral or liposomal genetic modification of the cardiomyocytes, with 24% providing reliable stimulation of the iPSC-CMs within 6 h and >80% within 24 h. Moreover, our data show that the spheroids can be frozen in liquid nitrogen for long-term storage and transportation, after which they can be deployed as a reagent on site for optical cardiac pacing. In all cases, optical stimulation was achieved at relatively low light levels (<0.15 mW/mm2) when 5 ms or longer pulses were used. Our results demonstrate a scalable, cost-effective method with a cryopreservable reagent to achieve contactless optical stimulation of cardiac cell constructs without genetically modifying the myocytes, that can be integrated in a robotics-amenable workflow for high-throughput drug testing.
Collapse
Affiliation(s)
- Christianne J Chua
- Cardiac Optogenetics & Optical Imaging Lab, Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Julie L Han
- Cardiac Optogenetics & Optical Imaging Lab, Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Weizhen Li
- Cardiac Optogenetics & Optical Imaging Lab, Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Wei Liu
- Cardiac Optogenetics & Optical Imaging Lab, Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Emilia Entcheva
- Cardiac Optogenetics & Optical Imaging Lab, Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| |
Collapse
|
76
|
Gruber A, Edri O, Huber I, Arbel G, Gepstein A, Shiti A, Shaheen N, Chorna S, Landesberg M, Gepstein L. Optogenetic modulation of cardiac action potential properties may prevent arrhythmogenesis in short and long QT syndromes. JCI Insight 2021; 6:e147470. [PMID: 34100384 PMCID: PMC8262308 DOI: 10.1172/jci.insight.147470] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022] Open
Abstract
Abnormal action potential (AP) properties, as occurs in long or short QT syndromes (LQTS and SQTS, respectively), can cause life-threatening arrhythmias. Optogenetics strategies, utilizing light-sensitive proteins, have emerged as experimental platforms for cardiac pacing, resynchronization, and defibrillation. We tested the hypothesis that similar optogenetic tools can modulate the cardiomyocyte's AP properties, as a potentially novel antiarrhythmic strategy. Healthy control and LQTS/SQTS patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were transduced to express the light-sensitive cationic channel channelrhodopsin-2 (ChR2) or the anionic-selective opsin, ACR2. Detailed patch-clamp, confocal-microscopy, and optical mapping studies evaluated the ability of spatiotemporally defined optogenetic protocols to modulate AP properties and prevent arrhythmogenesis in the hiPSC-CMs cell/tissue models. Depending on illumination timing, light-induced ChR2 activation induced robust prolongation or mild shortening of AP duration (APD), while ACR2 activation allowed effective APD shortening. Fine-tuning these approaches allowed for the normalization of pathological AP properties and suppression of arrhythmogenicity in the LQTS/SQTS hiPSC-CM cellular models. We next established a SQTS-hiPSC-CMs-based tissue model of reentrant-arrhythmias using optogenetic cross-field stimulation. An APD-modulating optogenetic protocol was then designed to dynamically prolong APD of the propagating wavefront, completely preventing arrhythmogenesis in this model. This work highlights the potential of optogenetics in studying repolarization abnormalities and in developing novel antiarrhythmic therapies.
Collapse
Affiliation(s)
- Amit Gruber
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Oded Edri
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Irit Huber
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Gil Arbel
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Amira Gepstein
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Assad Shiti
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Naim Shaheen
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Snizhana Chorna
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Michal Landesberg
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel
| | - Lior Gepstein
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine, the Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel, Haifa, Israel.,Cardiology Department, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
77
|
Zakharov A, Dasbiswas K. Mechanochemical induction of wrinkling morphogenesis on elastic shells. SOFT MATTER 2021; 17:4738-4750. [PMID: 33978668 DOI: 10.1039/d1sm00003a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Morphogenetic dynamics of tissue sheets require coordinated cell shape changes regulated by global patterning of mechanical forces. Inspired by such biological phenomena, we propose a minimal mechanochemical model based on the notion that cell shape changes are induced by diffusible biomolecules that influence tissue contractility in a concentration-dependent manner - and whose concentration is in turn affected by the macroscopic tissue shape. We perform computational simulations of thin shell elastic dynamics to reveal propagating chemical and three-dimensional deformation patterns arising due to a sequence of buckling instabilities. Depending on the concentration threshold that actuates cell shape change, we find qualitatively different patterns. The mechanochemically coupled patterning dynamics are distinct from those driven by purely mechanical or purely chemical factors, and emerge even without diffusion. Using numerical simulations and theoretical arguments, we analyze the elastic instabilities that result from our model and provide simple scaling laws to identify wrinkling morphologies.
Collapse
Affiliation(s)
- Andrei Zakharov
- Department of Physics, University of California, Merced, Merced, CA 95343, USA.
| | - Kinjal Dasbiswas
- Department of Physics, University of California, Merced, Merced, CA 95343, USA.
| |
Collapse
|
78
|
VanGordon MR, Prignano LA, Dempski RE, Rick SW, Rempe SB. Channelrhodopsin C1C2: Photocycle kinetics and interactions near the central gate. Biophys J 2021; 120:1835-1845. [PMID: 33705762 PMCID: PMC8204341 DOI: 10.1016/j.bpj.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/27/2022] Open
Abstract
Channelrhodopsins (ChR) are light-sensitive cation channels used in optogenetics, a technique that applies light to control cells (e.g., neurons) that have been modified genetically to express those channels. Although mutations are known to affect pore kinetics, little is known about how mutations induce changes at the molecular scale. To address this issue, we first measured channel opening and closing rates of a ChR chimera (C1C2) and selected variants (N297D, N297V, and V125L). Then, we used atomistic simulations to correlate those rates with changes in pore structure, hydration, and chemical interactions among key gating residues of C1C2 in both closed and open states. Overall, the experimental results show that C1C2 and its mutants do not behave like ChR2 or its analogous variants, except V125L, making C1C2 a unique channel. Our atomistic simulations confirmed that opening of the channel and initial hydration of the gating regions between helices I, II, III, and VII of the channel occurs with 1) the presence of 13-cis retinal; 2) deprotonation of a glutamic acid gating residue, E129; and 3) subsequent weakening of the central gate hydrogen bond between the same glutamic acid E129 and asparagine N297 in the central region of the pore. Also, an aspartate (D292) is the unambiguous primary proton acceptor for the retinal Schiff base in the hydrated channel.
Collapse
Affiliation(s)
- Monika R VanGordon
- Department of Chemistry, University of New Orleans, New Orleans, Louisiana
| | - Lindsey A Prignano
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Robert E Dempski
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts
| | - Steven W Rick
- Department of Chemistry, University of New Orleans, New Orleans, Louisiana
| | - Susan B Rempe
- Sandia National Laboratories, Albuquerque, New Mexico.
| |
Collapse
|
79
|
Ingles-Prieto A, Furthmann N, Crossman SH, Tichy AM, Hoyer N, Petersen M, Zheden V, Biebl J, Reichhart E, Gyoergy A, Siekhaus DE, Soba P, Winklhofer KF, Janovjak H. Optogenetic delivery of trophic signals in a genetic model of Parkinson's disease. PLoS Genet 2021; 17:e1009479. [PMID: 33857132 PMCID: PMC8049241 DOI: 10.1371/journal.pgen.1009479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
Optogenetics has been harnessed to shed new mechanistic light on current and future therapeutic strategies. This has been to date achieved by the regulation of ion flow and electrical signals in neuronal cells and neural circuits that are known to be affected by disease. In contrast, the optogenetic delivery of trophic biochemical signals, which support cell survival and are implicated in degenerative disorders, has never been demonstrated in an animal model of disease. Here, we reengineered the human and Drosophila melanogaster REarranged during Transfection (hRET and dRET) receptors to be activated by light, creating one-component optogenetic tools termed Opto-hRET and Opto-dRET. Upon blue light stimulation, these receptors robustly induced the MAPK/ERK proliferative signaling pathway in cultured cells. In PINK1B9 flies that exhibit loss of PTEN-induced putative kinase 1 (PINK1), a kinase associated with familial Parkinson's disease (PD), light activation of Opto-dRET suppressed mitochondrial defects, tissue degeneration and behavioral deficits. In human cells with PINK1 loss-of-function, mitochondrial fragmentation was rescued using Opto-dRET via the PI3K/NF-кB pathway. Our results demonstrate that a light-activated receptor can ameliorate disease hallmarks in a genetic model of PD. The optogenetic delivery of trophic signals is cell type-specific and reversible and thus has the potential to inspire novel strategies towards a spatio-temporal regulation of tissue repair.
Collapse
Affiliation(s)
- Alvaro Ingles-Prieto
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Nikolas Furthmann
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Samuel H. Crossman
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Alexandra-Madelaine Tichy
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Nina Hoyer
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meike Petersen
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Zheden
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Julia Biebl
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Eva Reichhart
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
| | - Attila Gyoergy
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Daria E. Siekhaus
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Peter Soba
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstanze F. Winklhofer
- Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Bochum, Germany
| | - Harald Janovjak
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, Australia
- European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Australia
- * E-mail:
| |
Collapse
|
80
|
Chen Z, Tsytsarev V, Finfrock YZ, Antipova OA, Cai Z, Arakawa H, Lischka FW, Hooks BM, Wilton R, Wang D, Liu Y, Gaitan B, Tao Y, Chen Y, Erzurumlu RS, Yang H, Rozhkova EA. Wireless Optogenetic Modulation of Cortical Neurons Enabled by Radioluminescent Nanoparticles. ACS NANO 2021; 15:5201-5208. [PMID: 33625219 DOI: 10.1021/acsnano.0c10436] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
While offering high-precision control of neural circuits, optogenetics is hampered by the necessity to implant fiber-optic waveguides in order to deliver photons to genetically engineered light-gated neurons in the brain. Unlike laser light, X-rays freely pass biological barriers. Here we show that radioluminescent Gd2(WO4)3:Eu nanoparticles, which absorb external X-rays energy and then downconvert it into optical photons with wavelengths of ∼610 nm, can be used for the transcranial stimulation of cortical neurons expressing red-shifted, ∼590-630 nm, channelrhodopsin ReaChR, thereby promoting optogenetic neural control to the practical implementation of minimally invasive wireless deep brain stimulation.
Collapse
Affiliation(s)
- Zhaowei Chen
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
- Institute of Food Safety and Environment Monitoring, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Vassiliy Tsytsarev
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Y Zou Finfrock
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
- Science Division, Canadian Light Source Inc., 44 Innovation Boulevard, Saskatoon, Saskatchewan S7N 2 V3, Canada
| | - Olga A Antipova
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Zhonghou Cai
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Fritz W Lischka
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814-4799, United States
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh, 3500 Terrace Street, Suite W1458, Pittsburgh, Pennsylvania 15213-2500, United States
| | - Rosemarie Wilton
- Biosciences, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Dongyi Wang
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yi Liu
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Brandon Gaitan
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yang Tao
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yu Chen
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Huanghao Yang
- Institute of Food Safety and Environment Monitoring, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Elena A Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| |
Collapse
|
81
|
Wang X, Cheng Y. Optical Manipulation of Perfused Mouse Heart Expressing Channelrhodopsin-2 in Rhythm Control. Methods Mol Biol 2021; 2191:377-390. [PMID: 32865755 DOI: 10.1007/978-1-0716-0830-2_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Optogenetics is a new approach using light intensity to modulate the electrical activity of excitable cells by the interaction of light-sensitive proteins. This method has been widely and enthusiastically utilized in some fields over the last decade. Localizing a photosensitive protein to a specific place in the membrane of cardiomyocytes at a specific time is essential for most biological processes. In this case, vectors are injected into the circulation to allow them to spread throughout the whole heart. The aim of this protocol is to perform different illumination modes with blue laser to investigate optical control of Langendorff-perfused mice hearts which were systematically injected with adeno-associated virus (AAV) for ChR2(H134R) gene transfer. Electrograms (EGs) and epicardium monophasic action potential (MAP) showed that ChR2 expression in the heart can be flexibly controlled by blue light across different illumination sites with corresponding triggered ectopic rhythm. Illumination intensity, pulse duration, and impulse frequency were associated with the light capture rate. Flexible control of the cardiac rhythm with optogenetics provides an innovative approach to cardiac research and therapy.
Collapse
Affiliation(s)
- Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China. .,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China.
| | - Yue Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.,Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| |
Collapse
|
82
|
Vogt M, Schulz B, Wagdi A, Lebert J, van Belle GJ, Christoph J, Bruegmann T, Patejdl R. Direct optogenetic stimulation of smooth muscle cells to control gastric contractility. Am J Cancer Res 2021; 11:5569-5584. [PMID: 33859764 PMCID: PMC8039938 DOI: 10.7150/thno.53883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
Rationale: Antral peristalsis is responsible for gastric emptying. Its failure is called gastroparesis and often caused by dysfunction of enteric neurons and interstitial cells of Cajal (ICC). Current treatment options, including gastric electrical stimulation, are non-satisfying and may improve symptoms but commonly fail to restore gastric emptying. Herein, we explore direct optogenetic stimulation of smooth muscle cells (SMC) via the light-gated non-selective cation channel Channelrhodopsin2 (ChR2) to control gastric motor function. Methods: We used a transgenic mouse model expressing ChR2 in fusion with eYFP under the control of the chicken-β-actin promoter. We performed patch clamp experiments to quantify light-induced currents in isolated SMC, Ca2+ imaging and isometric force measurements of antral smooth muscle strips as well as pressure recordings of intact stomachs to evaluate contractile responses. Light-induced propulsion of gastric contents from the isolated stomach preparation was quantified in video recordings. We furthermore tested optogenetic stimulation in a gastroparesis model induced by neuronal- and ICC-specific damage through methylene blue photo-toxicity. Results: In the stomachs, eYFP signals were restricted to SMC in which blue light (460 nm) induced inward currents typical for ChR2. These depolarizing currents led to contractions in antral smooth muscle strips that were stronger than those triggered by supramaximal electrical field stimulation and comparable to those evoked by global depolarization with high K+ concentration. In the intact stomach, panoramic illumination efficiently increased intragastric pressure achieving 239±46% (n=6) of the pressure induced by electrical field stimulation and triggered gastric transport. Within the gastroparesis model, electric field stimulation completely failed but light still efficiently generated pressure waves. Conclusions: We demonstrate direct optogenetic stimulation of SMC to control gastric contractility. This completely new approach could allow for the restoration of motility in gastroparesis in the future.
Collapse
|
83
|
Li J, Li H, Rao P, Luo J, Wang X, Wang L. Shining light on cardiac electrophysiology: From detection to intervention, from basic research to translational applications. Life Sci 2021; 274:119357. [PMID: 33737082 DOI: 10.1016/j.lfs.2021.119357] [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: 12/05/2020] [Revised: 03/01/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Cardiac arrhythmias are an important group of cardiovascular diseases, which can occur alone or in association with other cardiovascular diseases. The development of cardiac arrhythmias cannot be separated from changes in cardiac electrophysiology, and the investigation and clarification of cardiac electrophysiological changes are beneficial for the treatment of cardiac arrhythmias. However, electrical energy-based pacemakers and defibrillators, which are widely used to treat arrhythmias, still have certain disadvantages. Thereby, optics promises to be used for optical manipulation and its use in biomedicine is increasing. Since visible light is readily absorbed and scattered in living tissues and tissue penetration is shallow, optical modulation for cells and tissues requires conversion media that convert light energy into bioelectrical activity. In this regard, fluorescent dyes, light-sensitive ion channels, and optical nanomaterials can assume this role, the corresponding optical mapping technology, optogenetics technology, and optical systems based on luminescent nanomaterials have been introduced into the research in cardiovascular field and are expected to be new tools for the study and treatment of cardiac arrhythmias. In addition, infrared and near-infrared light has strong tissue penetration, which is one of the excellent options of external trigger for achieving optical modulation, and is also widely used in the study of optical modulation of biological activities. Here, the advantages of optical applications are summarized, the research progresses and emerging applications of optical-based technologies as detection and intervention tools for cardiac electrophysiological are highlighted. Moreover, the prospects for future applications of optics in clinical diagnosis and treatment are discussed.
Collapse
Affiliation(s)
- Jianyi Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Haitao Li
- Department of Cardiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, PR China
| | - Panpan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Junmiao Luo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China.
| |
Collapse
|
84
|
Observing and Manipulating Cell-Specific Cardiac Function with Light. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33398827 DOI: 10.1007/978-981-15-8763-4_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
The heart is a complex multicellular organ comprising both cardiomyocytes (CM), which make up the majority of the cardiac volume, and non-myocytes (NM), which represent the majority of cardiac cells. CM drive the pumping action of the heart, triggered via rhythmic electrical activity. NM, on the other hand, have many essential functions including generating extracellular matrix, regulating CM activity, and aiding in repair following injury. NM include neurons and interstitial, immune, and endothelial cells. Understanding the role of specific cell types and their interactions with one another may be key to developing new therapies with minimal side effects to treat cardiac disease. However, assessing cell-type-specific behavior in situ using standard techniques is challenging. Optogenetics enables population-specific observation and control, facilitating studies into the role of specific cell types and subtypes. Optogenetic models targeting the most important cardiac cell types have been generated and used to investigate non-canonical roles of those cell populations, e.g., to better understand how cardiac pacing occurs and to assess potential translational possibilities of optogenetics. So far, cardiac optogenetic studies have primarily focused on validating models and tools in the healthy heart. The field is now in a position where animal models and tools should be utilized to improve our understanding of the complex heterocellular nature of the heart, how this changes in disease, and from there to enable the development of cell-specific therapies and improved treatments.
Collapse
|
85
|
Ge A, Hu L, Fan J, Ge M, Wang X, Wang S, Feng X, Du W, Liu BF. A low-cost microfluidic platform coupled with light emitting diode for optogenetic analysis of neuronal response in C. elegans. Talanta 2021; 223:121646. [PMID: 33303134 DOI: 10.1016/j.talanta.2020.121646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 11/24/2022]
Abstract
Optogenetic method is widely used for dissecting the neuronal function and connectivity in a specific neural circuit, which can help understanding how the animal process information and generate behavior. The nematode C. elegans has a simple but complete nervous system, making it an attractive model to study the dynamics signals of neural circuits. However, in vivo analysis on neural circuits usually rely on the complex and expensive optical equipment to allow optogenetic stimulating the neuron while recording its activities in such a freely moving animal. Hence, in this paper we reported a portable optofluidic platform that works based on optical fiber illumination and functional imaging for worm optogenetic manipulation. A light beam from LED laser pen crossing the 3D-printed optical fiber channel is used to activate the neurons specific-expressed with light sensitive proteins ChR-2. The imaging light path is perpendicular to the stimulation light, which allows activating neuron precisely and measuring cellular signals simultaneously. By using such an easy-to-assemble device, optical stimulation of the specific neurons and detection of dynamic calcium responses of other neurons could be proceeded simultaneously. Thus, the developed microfluidic platform puts forward a simple, rapid and low-cost strategy for further neural circuits studies.
Collapse
Affiliation(s)
- Anle Ge
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Single Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
| | - Liang Hu
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - JiaXing Fan
- Department of Urology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Minghai Ge
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xixian Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Single Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
| | - Shanshan Wang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaojun Feng
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wei Du
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics - Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
86
|
Hussaini S, Venkatesan V, Biasci V, Romero Sepúlveda JM, Quiñonez Uribe RA, Sacconi L, Bub G, Richter C, Krinski V, Parlitz U, Majumder R, Luther S. Drift and termination of spiral waves in optogenetically modified cardiac tissue at sub-threshold illumination. eLife 2021; 10:59954. [PMID: 33502313 PMCID: PMC7840178 DOI: 10.7554/elife.59954] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/28/2020] [Indexed: 01/01/2023] Open
Abstract
The development of new approaches to control cardiac arrhythmias requires a deep understanding of spiral wave dynamics. Optogenetics offers new possibilities for this. Preliminary experiments show that sub-threshold illumination affects electrical wave propagation in the mouse heart. However, a systematic exploration of these effects is technically challenging. Here, we use state-of-the-art computer models to study the dynamic control of spiral waves in a two-dimensional model of the adult mouse ventricle, using stationary and non-stationary patterns of sub-threshold illumination. Our results indicate a light-intensity-dependent increase in cellular resting membrane potentials, which together with diffusive cell-cell coupling leads to the development of spatial voltage gradients over differently illuminated areas. A spiral wave drifts along the positive gradient. These gradients can be strategically applied to ensure drift-induced termination of a spiral wave, both in optogenetics and in conventional methods of electrical defibrillation.
Collapse
Affiliation(s)
- Sayedeh Hussaini
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,Institute for the Dynamics of Complex Systems, Goettingen University, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany
| | - Vishalini Venkatesan
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,University Medical Center Goettingen, Clinic of Cardiology and Pneumology, Goettingen, Germany
| | - Valentina Biasci
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino (FI), Italy.,Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Raul A Quiñonez Uribe
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany
| | - Leonardo Sacconi
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino (FI), Italy.,Institute for Experimental Cardiovascular Medicine, University of Freiburg, Freiburg, Germany.,National Institute of Optics, National Research Council, Florence, Italy
| | - Gil Bub
- Department of Physiology, MGill University, Montreal, Canada
| | - Claudia Richter
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany.,University Medical Center Goettingen, Clinic of Cardiology and Pneumology, Goettingen, Germany
| | - Valentin Krinski
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany.,INPHYNI, CNRS, Sophia Antipolis, Paris, France
| | - Ulrich Parlitz
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,Institute for the Dynamics of Complex Systems, Goettingen University, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany
| | - Rupamanjari Majumder
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany
| | - Stefan Luther
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Goettingen, Germany.,Institute for the Dynamics of Complex Systems, Goettingen University, Goettingen, Germany.,German Center for Cardiovascular Research, Partner Site Goettingen, Goettingen, Germany.,University Medical Center Goettingen, Institute of Pharmacology and Toxicology, Goettingen, Germany
| |
Collapse
|
87
|
Abstract
Computer simulations show how low-intensity illumination can be used to terminate cardiac arrhythmias.
Collapse
Affiliation(s)
- Eike M Wülfers
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad KrozingenBad KrozingenGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg – Bad KrozingenBad KrozingenGermany
- Faculty of Medicine, University of FreiburgFreiburgGermany
| |
Collapse
|
88
|
Abstract
Molecular dynamics (MD) simulations have been successfully used for modeling dynamic behavior of biologically relevant systems, such as ion channels in representative environments to decode protein structure-function relationships. Protocol presented here describes steps for generating input files and modeling a monomer of transmembrane cation channel, channelrhodopsin chimera (C1C2), in representative environment of 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) planar lipid bilayer, TIP3P water and ions (Na+ and Cl-) using molecular dynamics package NAMD, molecular graphics/analysis tool VMD, and other relevant tools. MD simulations of C1C2 were performed at 303.15 K and in constant particle number, isothermal-isobaric (NpT) ensemble. The results of modeling have helped understand how key interactions in the center of the C1C2 channel contribute to channel gating and subsequent solvent transport across the membrane.
Collapse
Affiliation(s)
- Monika R VanGordon
- Department of Chemistry, University of New Orleans, New Orleans, LA, USA.
| |
Collapse
|
89
|
Fernández MC, Kopton RA, Simon-Chica A, Madl J, Hilgendorf I, Zgierski-Johnston CM, Schneider-Warme F. Channelrhodopsins for Cell-Type Specific Illumination of Cardiac Electrophysiology. Methods Mol Biol 2021; 2191:287-307. [PMID: 32865751 DOI: 10.1007/978-1-0716-0830-2_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Optogenetic approaches have evolved as potent means to investigate cardiac electrophysiology, with research ranging from the study of arrhythmia mechanisms to effects of cardiac innervation and heterocellular structural and functional interactions, both in healthy and diseased myocardium. Most commonly, these studies use channelrhodopsin-2 (ChR2)-expressing murine models that enable light-activated depolarization of the target cell population. However, each newly generated mouse line requires thorough characterization, as cell-type specific ChR2 expression cannot be taken for granted, and the electrophysiological response of its activation in the target cell should be evaluated. In this chapter, we describe detailed protocols for assessing ChR2 specificity using immunohistochemistry, isolation of specific cell populations to analyze electrophysiological effects of ChR2 activation with the patch-clamp technique, and whole-heart experiments to assess in situ effects of optical stimulation.
Collapse
Affiliation(s)
- Marbely C Fernández
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ramona A Kopton
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ana Simon-Chica
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Josef Madl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiology I, University Heart Center Freiburg-Bad Krozingen, Freiburg, Germany
| | - Callum M Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Freiburg, Germany. .,Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
90
|
Asano T, Teh DBL, Yawo H. Application of Optogenetics for Muscle Cells and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:359-375. [PMID: 33398826 DOI: 10.1007/978-981-15-8763-4_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This chapter describes the current progress of basic research, and potential therapeutic applications primarily focused on the optical manipulation of muscle cells and neural stem cells using microbial rhodopsin as a light-sensitive molecule. Since the contractions of skeletal, cardiac, and smooth muscle cells are mainly regulated through their membrane potential, several studies have been demonstrated to up- or downregulate the muscle contraction directly or indirectly using optogenetic actuators or silencers with defined stimulation patterns and intensities. Light-dependent oscillation of membrane potential also facilitates the maturation of myocytes with the development of T tubules and sarcomere structures, tandem arrays of minimum contractile units consists of contractile proteins and cytoskeletal proteins. Optogenetics has been applied to various stem cells and multipotent/pluripotent cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) to generate light-sensitive neurons and to facilitate neuroscience. The chronic optical stimulation of the channelrhodopsin-expressing neural stem cells facilitates their neural differentiation. There are potential therapeutic applications of optogenetics in cardiac pacemaking, muscle regeneration/maintenance, locomotion recovery for the treatment of muscle paralysis due to motor neuron diseases such as amyotrophic lateral sclerosis (ALS). Optogenetics would also facilitate maturation, network integration of grafted neurons, and improve the microenvironment around them when applied to stem cells.
Collapse
Affiliation(s)
- Toshifumi Asano
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Daniel Boon Loong Teh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiromu Yawo
- The Institute for Solid State Physics, The University of Tokyo, Kashiwa, Japan.
| |
Collapse
|
91
|
Ganji E, Chan CS, Ward CW, Killian ML. Optogenetic activation of muscle contraction in vivo. Connect Tissue Res 2021; 62:15-23. [PMID: 32777957 PMCID: PMC7718400 DOI: 10.1080/03008207.2020.1798943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/15/2020] [Indexed: 02/03/2023]
Abstract
Purpose: Optogenetics is an emerging alternative to traditional electrical stimulation to initiate action potentials in activatable cells both ex vivo and in vivo. Optogenetics has been commonly used in mammalian neurons and more recently, it has been adapted for activation of cardiomyocytes and skeletal muscle. Therefore, the aim of this study was to evaluate the stimulation feasibility and sustain isometric muscle contraction and limit decay for an extended period of time (1s), using non-invasive transdermal light activation of skeletal muscle (triceps surae) in vivo. MATERIALS AND METHODS We used inducible Cre recombination to target expression of Channelrhodopsin-2 (ChR2(H134R)-EYFP) in skeletal muscle (Acta1-Cre) in mice. Fluorescent imaging confirmed that ChR2 expression is localized in skeletal muscle and does not have specific expression in sciatic nerve branch, therefore, allowing for non-nerve mediated optical stimulation of skeletal muscle. We induced muscle contraction using transdermal exposure to blue light and selected 10 Hz stimulation after controlled optimization experiments to sustain prolonged muscle contraction. RESULTS Increasing the stimulation frequency from 10 Hz to 40 Hz increased the muscle contraction decay during prolonged 1s stimulation, highlighting frequency dependency and importance of membrane repolarization for effective light activation. Finally, we showed that optimized pulsed optogenetic stimulation of 10 Hz resulted in comparable ankle torque and contractile functionality to that of electrical stimulation. CONCLUSIONS Our results demonstrate the feasibility and repeatability of non-invasive optogenetic stimulation of muscle in vivo and highlight optogenetic stimulation as a powerful tool for non-invasive in vivo direct activation of skeletal muscle.
Collapse
Affiliation(s)
- Elahe Ganji
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
- Department of Orthopaedic Surgery, Michigan Medicine, Ann Arbor, Michigan, 48109
| | - C. Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Christopher W. Ward
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Megan L. Killian
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
- Department of Orthopaedic Surgery, Michigan Medicine, Ann Arbor, Michigan, 48109
| |
Collapse
|
92
|
Dwenger M, Kowalski WJ, Masumoto H, Nakane T, Keller BB. Chronic Optogenetic Pacing of Human-Induced Pluripotent Stem Cell-Derived Engineered Cardiac Tissues. Methods Mol Biol 2021; 2191:151-169. [PMID: 32865744 DOI: 10.1007/978-1-0716-0830-2_10] [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] [Indexed: 12/12/2022]
Abstract
The delivery of cells into damaged myocardium induces limited cardiac regeneration due to extensive cell death. In an effort to limit cell death, our lab formulates three-dimensional matrices as a delivery system for cell therapy. Our primary work has been focused on the formation of engineered cardiac tissues (ECTs) from human-induced pluripotent stem cell-derived engineered cardiac cells. However, ECT immaturity hinders ability to fully recover damaged myocardium. Various conditioning regimens such as mechanical stretch and/or electric pacing have been used to activate maturation pathways. To improve ECT maturity, we use non-contacting chronic light stimulation using heterologously expressed light-sensitive channelrhodopsin ion channels. We transduce ECTs with an AAV packaged channelrhodopsin and chronically optically pace (C-OP) ECTs for 1 week above the intrinsic beat rate, resulting in increased ECT electrophysiological properties.
Collapse
Affiliation(s)
- Marc Dwenger
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - William J Kowalski
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Hidetoshi Masumoto
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.,Department of CV Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takeichiro Nakane
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of CV Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Bradley B Keller
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA. .,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, USA. .,Cincinnati Children's Heart Institute, Cincinnati Children's Hospital Medical Center, Louisville, KY, USA.
| |
Collapse
|
93
|
Diaz Vera D, Soucy JR, Lee A, Koppes RA, Koppes AN. Light irradiation of peripheral nerve cells: Wavelength impacts primary sensory neuron outgrowth in vitro. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 215:112105. [PMID: 33406470 DOI: 10.1016/j.jphotobiol.2020.112105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022]
Abstract
The expansion of optogenetics via the development and application of new opsins has opened a new world of possibilities as a research and therapeutic tool. Nevertheless, it has also raised questions about the innocuity of using light irradiation on tissues and cells such as those from the Peripheral Nervous System (PNS). Thus, to investigate the potential of PNS being affected by optogenetic light irradiation, rat dorsal root ganglion neurons and Schwann cells were isolated and their response to light irradiation examined in vitro. Light irradiation was delivered as millisecond pulses at wavelengths in the visible spectrum between 627 and 470 nm, with doses ranging between 4.5 and 18 J/cm2 at an irradiance value of 1 mW/mm2. Results show that compared to cultures kept in dark conditions, light irradiation at 470 nm reduced neurite outgrowth in dissociated dorsal root neurons in a dose dependent manner while higher wavelengths had no effect on neuron morphology. Although neurite outgrowth was limited by light irradiation, no signs of cell death or apoptosis were found. On the other hand, peripheral glia, Schwann cells, were insensitive to light irradiation with metabolism, proliferation, and RNA levels of transcription factors c-Jun and krox-20 remaining unaltered following stimulation. As the fields of photostimulation and optogenetics expand, these results indicate the need for consideration to cell type response and stimulation parameters for applications in vitro and further investigation on specific mechanisms driving response.
Collapse
Affiliation(s)
- David Diaz Vera
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | - Jonathan R Soucy
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | - Audrey Lee
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA; Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
94
|
Abstract
The electromechanical function of the heart involves complex, coordinated activity over time and space. Life-threatening cardiac arrhythmias arise from asynchrony in these space-time events; therefore, therapies for prevention and treatment require fundamental understanding and the ability to visualize, perturb and control cardiac activity. Optogenetics combines optical and molecular biology (genetic) approaches for light-enabled sensing and actuation of electrical activity with unprecedented spatiotemporal resolution and parallelism. The year 2020 marks a decade of developments in cardiac optogenetics since this technology was adopted from neuroscience and applied to the heart. In this Review, we appraise a decade of advances that define near-term (immediate) translation based on all-optical electrophysiology, including high-throughput screening, cardiotoxicity testing and personalized medicine assays, and long-term (aspirational) prospects for clinical translation of cardiac optogenetics, including new optical therapies for rhythm control. The main translational opportunities and challenges for optogenetics to be fully embraced in cardiology are also discussed.
Collapse
|
95
|
Johannsmeier S, Wenzel J, Torres-Mapa ML, Junge S, Sasse P, Stockhausen JD, Ripken T, Heinemann D, Heisterkamp A. Light-cell interactions in depth-resolved optogenetics. BIOMEDICAL OPTICS EXPRESS 2020; 11:6536-6550. [PMID: 33282507 PMCID: PMC7687973 DOI: 10.1364/boe.404388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 06/12/2023]
Abstract
Light as a tool in medical therapy and biological research has been studied extensively and its application is subject to continuous improvement. However, safe and efficient application of light-based methods in photomedicine or optogenetics requires knowledge about the optical properties of the target tissue as well as the response characteristics of the stimulated cells. Here, we used tissue phantoms and a heart-like light-sensitive cell line to investigate optogenetic stimulation through tissue layers. The input power necessary for successful stimulation could be described as a function of phantom thickness. A model of light transmission through the tissue phantoms gives insights into the expected stimulation efficiency. Cell-type specific effects are identified that result in deviations of the stimulation threshold from the modelled predictions. This study provides insights into the complex interplay between light, tissue and cells during deep-tissue optogenetics. It can serve as an orientation for safe implementation of light-based methods in vivo.
Collapse
Affiliation(s)
- Sonja Johannsmeier
- Industrial and Biomedical Optics Department, Laser Zentrum Hannover e.V., Hollerithallee 8, 30419 Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| | - Johannes Wenzel
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz Universität Hannover, Welfengarten 1, 30167 Hannover, Germany
| | - Maria L. Torres-Mapa
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz Universität Hannover, Welfengarten 1, 30167 Hannover, Germany
| | - Sebastian Junge
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz Universität Hannover, Welfengarten 1, 30167 Hannover, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Nussallee 11, 53115 Bonn, Germany
| | - Joshua D. Stockhausen
- Institute of Physiology I, Medical Faculty, University of Bonn, Nussallee 11, 53115 Bonn, Germany
| | - Tammo Ripken
- Industrial and Biomedical Optics Department, Laser Zentrum Hannover e.V., Hollerithallee 8, 30419 Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
| | - Dag Heinemann
- Industrial and Biomedical Optics Department, Laser Zentrum Hannover e.V., Hollerithallee 8, 30419 Hannover, Germany
- Department of Phytophotonics, Institute of Horticultural Production Systems and Hannover Centre for Optical Technologies (HOT), Gottfried Wilhelm Leibniz University Hannover, 30419 Hannover, Germany
| | - Alexander Heisterkamp
- Industrial and Biomedical Optics Department, Laser Zentrum Hannover e.V., Hollerithallee 8, 30419 Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, Stadtfelddamm 34, 30625 Hannover, Germany
- Institute of Quantum Optics, Gottfried Wilhelm Leibniz Universität Hannover, Welfengarten 1, 30167 Hannover, Germany
| |
Collapse
|
96
|
Cardiac Optogenetics in Atrial Fibrillation: Current Challenges and Future Opportunities. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8814092. [PMID: 33195698 PMCID: PMC7641281 DOI: 10.1155/2020/8814092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/07/2020] [Indexed: 12/23/2022]
Abstract
Although rarely life-threatening on short term, atrial fibrillation leads to increased mortality and decreased quality of life through its complications, including heart failure and stroke. Recent studies highlight the benefits of maintaining sinus rhythm. However, pharmacological long-term rhythm control strategies may be shadowed by associated proarrhythmic effects. At the same time, electrical cardioversion is limited to hospitals, while catheter ablation therapy, although effective, is invasive and is dedicated to specific patients, usually with low amounts of atrial fibrosis (preferably Utah I-II). Cardiac optogenetics allows influencing the heart's electrical activity by applying specific wavelength light pulses to previously engineered cardiomyocytes into expressing microbial derived light-sensitive proteins called opsins. The resulting ion influx may give rise to either hyperpolarizing or depolarizing currents, thus offering a therapeutic potential in cardiac electrophysiology, including pacing, resynchronization, and arrhythmia termination. Optogenetic atrial fibrillation cardioversion might be achieved by inducing a conduction block or filling of the excitable gap. The authors agree that transmural opsin expression and appropriate illumination with an exposure time longer than the arrhythmia cycle length are necessary to achieve successful arrhythmia termination. However, the efficiency and safety of biological cardioversion in humans remain to be seen, as well as side effects such as immune reactions and loss of opsin expression. The possibility of delivering pain-free shocks with out-of-hospital biological cardioversion is tempting; however, there are several issues that need to be addressed first: applicability and safety in humans, long-term behaviour, anticoagulation requirements, and fibrosis interactions.
Collapse
|
97
|
Li J, Wang L, Luo J, Li H, Rao P, Cheng Y, Wang X, Huang C. Optical capture and defibrillation in rats with monocrotaline-induced myocardial fibrosis 1 year after a single intravenous injection of adeno-associated virus channelrhodopsin-2. Heart Rhythm 2020; 18:109-117. [PMID: 32781160 DOI: 10.1016/j.hrthm.2020.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/28/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Optogenetics uses light to regulate cardiac rhythms and terminate malignant arrhythmias. OBJECTIVE The purpose of this study was to investigate the long-term validity of optical capture properties based on virus-transfected channelrhodopsin-2 (ChR2) and evaluate the effects of optogenetic-based defibrillation in an in vivo rat model of myocardial fibrosis enhanced by monocrotaline (MCT). METHODS Fifteen infant rats received jugular vein injection of adeno-associated virus (AAV). After 8 weeks, 5 rats were randomly selected to verify the effectiveness ChR2 transfection. The remaining rats were administered MCT at 11 months. Four weeks after MCT, the availability of 473-nm blue light to capture heart rhythm in these rats was verified again. Ventricular tachycardia (VT) and ventricular fibrillation (VF) were induced by burst stimulation on the basis of enhanced myocardial fibrosis, and the termination effects of the optical manipulation were tested. RESULTS Eight weeks after AAV injection, there was ChR2 expression throughout the ventricular myocardium as reflected by both fluorescence imaging and optical pacing. Four weeks after MCT, significant myocardial fibrosis was achieved. Light could still trigger the corresponding ectopic heart rhythm, and the pulse width and illumination area could affect the light capture rate. VT/VF was induced successfully in 1-year-observation rats, and the rate of termination of VT/VF under light was much higher than that of spontaneous termination. CONCLUSION Viral ChR2 transfection can play a long-term role in the rat heart, and light can successfully regulate heart rhythm and defibrillate after cardiac fibrosis.
Collapse
Affiliation(s)
- Jianyi Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Junmiao Luo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Haitao Li
- Department of Cardiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| | - Panpan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Yue Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China.
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China; Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China; Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China.
| |
Collapse
|
98
|
A non-invasive far-red light-induced split-Cre recombinase system for controllable genome engineering in mice. Nat Commun 2020; 11:3708. [PMID: 32709899 PMCID: PMC7381682 DOI: 10.1038/s41467-020-17530-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
The Cre-loxP recombination system is a powerful tool for genetic manipulation. However, there are widely recognized limitations with chemically inducible Cre-loxP systems, and the UV and blue-light induced systems have phototoxicity and minimal capacity for deep tissue penetration. Here, we develop a far-red light-induced split Cre-loxP system (FISC system) based on a bacteriophytochrome optogenetic system and split-Cre recombinase, enabling optogenetical regulation of genome engineering in vivo solely by utilizing a far-red light (FRL). The FISC system exhibits low background and no detectable photocytotoxicity, while offering efficient FRL-induced DNA recombination. Our in vivo studies showcase the strong organ-penetration capacity of FISC system, markedly outperforming two blue-light-based Cre systems for recombination induction in the liver. Demonstrating its strong clinical relevance, we successfully deploy a FISC system using adeno-associated virus (AAV) delivery. Thus, the FISC system expands the optogenetic toolbox for DNA recombination to achieve spatiotemporally controlled, non-invasive genome engineering in living systems. Current light-inducible Cre-loxP systems have minimal capacity for deep tissue penetration. Here, the authors present a far-red light-induced split Cre-loxP system for in vivo genome engineering.
Collapse
|
99
|
Burton RAB, Tomek J, Ambrosi CM, Larsen HE, Sharkey AR, Capel RA, Corbett AD, Bilton S, Klimas A, Stephens G, Cremer M, Bose SJ, Li D, Gallone G, Herring N, Mann EO, Kumar A, Kramer H, Entcheva E, Paterson DJ, Bub G. Optical Interrogation of Sympathetic Neuronal Effects on Macroscopic Cardiomyocyte Network Dynamics. iScience 2020; 23:101334. [PMID: 32674058 PMCID: PMC7363704 DOI: 10.1016/j.isci.2020.101334] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiac stimulation via sympathetic neurons can potentially trigger arrhythmias. We present approaches to study neuron-cardiomyocyte interactions involving optogenetic selective probing and all-optical electrophysiology to measure activity in an automated fashion. Here we demonstrate the utility of optical interrogation of sympathetic neurons and their effects on macroscopic cardiomyocyte network dynamics to address research targets such as the effects of adrenergic stimulation via the release of neurotransmitters, the effect of neuronal numbers on cardiac behavior, and the applicability of optogenetics in mechanistic in vitro studies. As arrhythmias are emergent behaviors that involve the coordinated activity of millions of cells, we image at macroscopic scales to capture complex dynamics. We show that neurons can both decrease and increase wave stability and re-entrant activity in culture depending on their induced activity-a finding that may help us understand the often conflicting results seen in experimental and clinical studies.
Collapse
Affiliation(s)
- Rebecca-Ann B Burton
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK; University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK.
| | - Jakub Tomek
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Christina M Ambrosi
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - Hege E Larsen
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Amy R Sharkey
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Rebecca A Capel
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | | | - Samuel Bilton
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Aleksandra Klimas
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - Guy Stephens
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Maegan Cremer
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | - Samuel J Bose
- University of Oxford, Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | - Dan Li
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Giuseppe Gallone
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK; Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany
| | - Neil Herring
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Edward O Mann
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Abhinav Kumar
- University of Oxford, Department of Biochemistry, Glycobiology Institute, Oxford, UK
| | - Holger Kramer
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Emilia Entcheva
- The George Washington University, Department of Biomedical Engineering, Washington, DC 20052, USA
| | - David J Paterson
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK
| | - Gil Bub
- University of Oxford, Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Research Excellence, Parks Road, Oxford OX1 3PT, UK; McGill University, Department of Physiology, McIntyre Medical Sciences Building, Room 1128, 3655 Promenade Sir William Osler, Montréal, QC H3G 1Y6, Canada.
| |
Collapse
|
100
|
Cheng Y, Li H, Wang L, Li J, Kang W, Rao P, Zhou F, Wang X, Huang C. Optogenetic approaches for termination of ventricular tachyarrhythmias after myocardial infarction in rats in vivo. JOURNAL OF BIOPHOTONICS 2020; 13:e202000003. [PMID: 32246523 DOI: 10.1002/jbio.202000003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Cardiac optogenetics facilitates the painless manipulation of the heart with optical energy and was recently shown to terminate ventricular tachycardia (VT) in explanted mice heart. This study aimed to evaluate the optogenetic-based termination of induced VT under ischemia in an open-chest rat model and to develop an optimal, optical-manipulation procedure. VT was induced by burst stimulation after ligation of the left anterior descending coronary artery, and the termination effects of the optical manipulation, including electrical anti-tachycardia pacing (ATP) and spontaneous recovery, were tested. Among different multisegment optical modes, four repeated illuminations of 1000 ms in duration with 1-second interval at a 20-times intensity threshold on the right ventricle achieved the highest termination rate of 86.14% ± 4.145%, higher than that achieved by ATP and spontaneous termination. We demonstrated that optogenetic-based cardioversion is feasible and effective in vivo, with the underlying mechanism involving the light-triggered, ChR2-induced depolarization of the illuminated myocardium, in turn generating an excitation that disrupts the preexisting reentrant wave front.
Collapse
Affiliation(s)
- Yue Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Haitao Li
- Department of Cardiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianyi Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Wen Kang
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Panpan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Fang Zhou
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
- Department of Cardiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| |
Collapse
|