101
|
Lemme M, Braren I, Prondzynski M, Aksehirlioglu B, Ulmer BM, Schulze ML, Ismaili D, Meyer C, Hansen A, Christ T, Lemoine MD, Eschenhagen T. Chronic intermittent tachypacing by an optogenetic approach induces arrhythmia vulnerability in human engineered heart tissue. Cardiovasc Res 2020; 116:1487-1499. [PMID: 31598634 PMCID: PMC7314638 DOI: 10.1093/cvr/cvz245] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/31/2019] [Accepted: 10/04/2019] [Indexed: 01/01/2023] Open
Abstract
AIMS Chronic tachypacing is commonly used in animals to induce cardiac dysfunction and to study mechanisms of heart failure and arrhythmogenesis. Human induced pluripotent stem cells (hiPSC) may replace animal models to overcome species differences and ethical problems. Here, 3D engineered heart tissue (EHT) was used to investigate the effect of chronic tachypacing on hiPSC-cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS To avoid cell toxicity by electrical pacing, we developed an optogenetic approach. EHTs were transduced with lentivirus expressing channelrhodopsin-2 (H134R) and stimulated by 15 s bursts of blue light pulses (0.3 mW/mm2, 30 ms, 3 Hz) separated by 15 s without pacing for 3 weeks. Chronic optical tachypacing did not affect contractile peak force, but induced faster contraction kinetics, shorter action potentials, and shorter effective refractory periods. This electrical remodelling increased vulnerability to tachycardia episodes upon electrical burst pacing. Lower calsequestrin 2 protein levels, faster diastolic depolarization (DD) and efficacy of JTV-519 (46% at 1 µmol/L) to terminate tachycardia indicate alterations of Ca2+ handling being part of the underlying mechanism. However, other antiarrhythmic compounds like flecainide (69% at 1 µmol/L) and E-4031 (100% at 1 µmol/L) were also effective, but not ivabradine (1 µmol/L) or SEA0400 (10 µmol/L). CONCLUSION We demonstrated a high vulnerability to tachycardia of optically tachypaced hiPSC-CMs in EHT and the effective termination by ryanodine receptor stabilization, sodium or hERG potassium channel inhibition. This new model might serve as a preclinical tool to test antiarrhythmic drugs increasing the insight in treating ventricular tachycardia.
Collapse
Affiliation(s)
- Marta Lemme
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ingke Braren
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Maksymilian Prondzynski
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, USA
| | - Bülent Aksehirlioglu
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Bärbel M Ulmer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Mirja L Schulze
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Djemail Ismaili
- Department of Cardiology-Electrophysiology, University Heart Center, 20246 Hamburg, Germany
| | - Christian Meyer
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology-Electrophysiology, University Heart Center, 20246 Hamburg, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Marc D Lemoine
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology-Electrophysiology, University Heart Center, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| |
Collapse
|
102
|
Boyle PM, Trayanova NA. Leave the light on: chronic optogenetic tachypacing of human engineered cardiac tissue constructs. Cardiovasc Res 2020; 116:1405-1406. [PMID: 32031599 DOI: 10.1093/cvr/cvaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Patrick M Boyle
- Department of Bioengineering and Institute for Stem Cell and Regenerative Medicine, University of Washington, Mailbox 355061, Seattle, WA 98195, USA
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, 216 Hackerman Hall, 3400 North Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
103
|
Men J, Li A, Jerwick J, Li Z, Tanzi RE, Zhou C. Non-invasive red-light optogenetic control of Drosophila cardiac function. Commun Biol 2020; 3:336. [PMID: 32601302 PMCID: PMC7324573 DOI: 10.1038/s42003-020-1065-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/03/2020] [Indexed: 02/03/2023] Open
Abstract
Drosophila is a powerful genetic model system for cardiovascular studies. Recently, optogenetic pacing tools have been developed to control Drosophila heart rhythm noninvasively with blue light, which has a limited penetration depth. Here we developed both a red-light sensitive opsin expressing Drosophila system and an integrated red-light stimulation and optical coherence microscopy (OCM) imaging system. We demonstrated noninvasive control of Drosophila cardiac rhythms using a single light source, including simulated tachycardia in ReaChR-expressing flies and bradycardia and cardiac arrest in halorhodopsin (NpHR)-expressing flies at multiple developmental stages. By using red excitation light, we were able to pace flies at higher efficiency and with lower power than with equivalent blue light excitation systems. The recovery dynamics after red-light stimulation of NpHR flies were observed and quantified. The combination of red-light stimulation, OCM imaging, and transgenic Drosophila systems provides a promising and easily manipulated research platform for noninvasive cardiac optogenetic studies. Men et al. develop an optogenetic pacing tool to control Drosophila heart rhythm noninvasively with red light. Using optical coherence microscopy imaging, they demonstrate effective light-induced tachypacing, bradypacing, and restorable cardiac arrest in transgenic fly models. This study provides a user-friendly research platform for noninvasive cardiac optogenetic studies.
Collapse
Affiliation(s)
- Jing Men
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA
| | - Airong Li
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Jason Jerwick
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA.,Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Zilong Li
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Chao Zhou
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA. .,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63105, USA. .,Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA, 18015, USA.
| |
Collapse
|
104
|
Sigalas C, Cremer M, Winbo A, Bose SJ, Ashton JL, Bub G, Montgomery JM, Burton RAB. Combining tissue engineering and optical imaging approaches to explore interactions along the neuro-cardiac axis. ROYAL SOCIETY OPEN SCIENCE 2020; 7:200265. [PMID: 32742694 PMCID: PMC7353978 DOI: 10.1098/rsos.200265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/27/2020] [Indexed: 05/05/2023]
Abstract
Interactions along the neuro-cardiac axis are being explored with regard to their involvement in cardiac diseases, including catecholaminergic polymorphic ventricular tachycardia, hypertension, atrial fibrillation, long QT syndrome and sudden death in epilepsy. Interrogation of the pathophysiology and pathogenesis of neuro-cardiac diseases in animal models present challenges resulting from species differences, phenotypic variation, developmental effects and limited availability of data relevant at both the tissue and cellular level. By contrast, tissue-engineered models containing cardiomyocytes and peripheral sympathetic and parasympathetic neurons afford characterization of cellular- and tissue-level behaviours while maintaining precise control over developmental conditions, cellular genotype and phenotype. Such approaches are uniquely suited to long-term, high-throughput characterization using optical recording techniques with the potential for increased translational benefit compared to more established techniques. Furthermore, tissue-engineered constructs provide an intermediary between whole animal/tissue experiments and in silico models. This paper reviews the advantages of tissue engineering methods of multiple cell types and optical imaging techniques for the characterization of neuro-cardiac diseases.
Collapse
Affiliation(s)
| | - Maegan Cremer
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Annika Winbo
- Department of Physiology, University of Auckland, Auckland, New Zealand
- Department of Paediatric and Congenital Cardiac Services, Starship Children's Hospital, Auckland, New Zealand
| | - Samuel J. Bose
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Jesse L. Ashton
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Gil Bub
- Department of Physiology, McGill University, Montreal, Canada
| | | | - Rebecca A. B. Burton
- Department of Pharmacology, University of Oxford, Oxford, UK
- Author for correspondence: Rebecca A. B. Burton e-mail:
| |
Collapse
|
105
|
Gundelach LA, Hüser MA, Beutner D, Ruther P, Bruegmann T. Towards the clinical translation of optogenetic skeletal muscle stimulation. Pflugers Arch 2020; 472:527-545. [PMID: 32415463 PMCID: PMC7239821 DOI: 10.1007/s00424-020-02387-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/05/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Abstract
Paralysis is a frequent phenomenon in many diseases, and to date, only functional electrical stimulation (FES) mediated via the innervating nerve can be employed to restore skeletal muscle function in patients. Despite recent progress, FES has several technical limitations and significant side effects. Optogenetic stimulation has been proposed as an alternative, as it may circumvent some of the disadvantages of FES enabling cell type–specific, spatially and temporally precise stimulation of cells expressing light-gated ion channels, commonly Channelrhodopsin2. Two distinct approaches for the restoration of skeletal muscle function with optogenetics have been demonstrated: indirect optogenetic stimulation through the innervating nerve similar to FES and direct optogenetic stimulation of the skeletal muscle. Although both approaches show great promise, both have their limitations and there are several general hurdles that need to be overcome for their translation into clinics. These include successful gene transfer, sustained optogenetic protein expression, and the creation of optically active implantable devices. Herein, a comprehensive summary of the underlying mechanisms of electrical and optogenetic approaches is provided. With this knowledge in mind, we substantiate a detailed discussion of the advantages and limitations of each method. Furthermore, the obstacles in the way of clinical translation of optogenetic stimulation are discussed, and suggestions on how they could be overcome are provided. Finally, four specific examples of pathologies demanding novel therapeutic measures are discussed with a focus on the likelihood of direct versus indirect optogenetic stimulation.
Collapse
Affiliation(s)
- Lili A Gundelach
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Marc A Hüser
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center, Göttingen, Germany
| | - Dirk Beutner
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center, Göttingen, Germany
| | - Patrick Ruther
- Microsystem Materials Laboratory, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Cluster of Excellence at the University of Freiburg, Freiburg, Germany
| | - Tobias Bruegmann
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany.
- DZHK e.V. (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
106
|
Rao P, Wang L, Cheng Y, Wang X, Li H, Zheng G, Li Z, Jiang C, Zhou Q, Huang C. Near-infrared light driven tissue-penetrating cardiac optogenetics via upconversion nanoparticles in vivo. BIOMEDICAL OPTICS EXPRESS 2020; 11:1401-1416. [PMID: 32206418 PMCID: PMC7075614 DOI: 10.1364/boe.381480] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/05/2019] [Accepted: 02/07/2020] [Indexed: 05/27/2023]
Abstract
This study determines whether near-infrared (NIR) light can drive tissue-penetrating cardiac optical control with upconversion luminescent materials. Adeno-associated virus (AAV) encoding channelrhodopsin-2 (ChR2) was injected intravenously to rats to achieve ChR2 expression in the heart. The upconversion nanoparticles (UCNP) NaYF4:Yb/Tm or upconversion microparticles (UCMP) NaYF4 to upconvert blue light were selected to fabricate freestanding polydimethylsiloxane films. These were attached on the ventricle and covered with muscle tissue. Additionally, a 980-nm NIR laser was programmed and illuminated on the film or the tissue. The NIR laser successfully captured ectopic paced rhythm in the heart, which displays similar manipulation characteristics to those triggered by blue light. Our results highlight the feasibility of tissue-penetration cardiac optogenetics by NIR and demonstrate the potential to use external optical manipulation for non-invasive or weakly invasive applications in cardiovascular diseases.
Collapse
Affiliation(s)
- Panpan Rao
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
- These authors contributed equally to this work
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- These authors contributed equally to this work
| | - Yue Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
- Co-corresponding authors
| | - Haitao Li
- Department of Cardiology, Hainan General Hospital, 570311, Haikou, China
| | - Guoxing Zheng
- School of Electronic Information, Wuhan University, 430072, Wuhan, China
- Co-corresponding authors
| | - Zile Li
- School of Electronic Information, Wuhan University, 430072, Wuhan, China
| | - Chan Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
| | - Qing Zhou
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, 430060, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, 430060, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, 430060, Wuhan, China
- Hubei Key Laboratory of Cardiology,430060, Wuhan, China
| |
Collapse
|
107
|
Joshi J, Rubart M, Zhu W. Optogenetics: Background, Methodological Advances and Potential Applications for Cardiovascular Research and Medicine. Front Bioeng Biotechnol 2020; 7:466. [PMID: 32064254 PMCID: PMC7000355 DOI: 10.3389/fbioe.2019.00466] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/19/2019] [Indexed: 12/27/2022] Open
Abstract
Optogenetics is an elegant approach of precisely controlling and monitoring the biological functions of a cell, group of cells, tissues, or organs with high temporal and spatial resolution by using optical system and genetic engineering technologies. The field evolved with the need to precisely control neurons and decipher neural circuity and has made great accomplishments in neuroscience. It also evolved in cardiovascular research almost a decade ago and has made considerable progress in both in vitro and in vivo animal studies. Thus, this review is written with an objective to provide information on the evolution, background, methodical advances, and potential scope of the field for cardiovascular research and medicine. We begin with a review of literatures on optogenetic proteins related to their origin, structure, types, mechanism of action, methods to improve their performance, and the delivery vehicles and methods to express such proteins on target cells and tissues for cardiovascular research. Next, we reviewed historical and recent literatures to demonstrate the scope of optogenetics for cardiovascular research and regenerative medicine and examined that cardiac optogenetics is vital in mimicking heart diseases, understanding the mechanisms of disease progression and also in introducing novel therapies to treat cardiac abnormalities, such as arrhythmias. We also reviewed optogenetics as promising tools in providing high-throughput data for cardiotoxicity screening in drug development and also in deciphering dynamic roles of signaling moieties in cell signaling. Finally, we put forth considerations on the need of scaling up of the optogenetic system, clinically relevant in vivo and in silico models, light attenuation issues, and concerns over the level, immune reactions, toxicity, and ectopic expression with opsin expression. Detailed investigations on such considerations would accelerate the translation of cardiac optogenetics from present in vitro and in vivo animal studies to clinical therapies.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Mayo Clinic, Phoenix, AZ, United States
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
108
|
Luo JW, Nikolic K, Degenaar P. Modelling Optogenetic Subthreshold Effects. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:6136-6140. [PMID: 31947244 DOI: 10.1109/embc.2019.8856664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We develop a system-level approach to modelling optogenetic-neurons firing behaviour in in-vivo conditions. This approach contains three sub-modules: 1) a Mie/Rayleigh scattering mode of light penetration in tissue; 2) a classic likelihood Poisson spiking train model; 3) a 4-state model of the Channelrhodopsin-2 (ChR2) channel added to a CA3 neuron Hodgkin-Huxley model. We first investigate opto-neurons lightto-spike mechanisms in an in-vivo model: the background noise (synaptic currents) play a dominant role in generating spikes rather than light intensities as for in-vitro conditions (Typically the required light intensity is less than 0.3 mW/mm2 for in-vivo). Then the spiking fidelity is analyzed for different background noise levels. Next, by combining light penetration profiles, we show how neuron firing rates decay as tissue distance increases, for a 2D dimensional cross-section. This preliminary data clearly demonstrate that at given light stimulation protocol, the maximum effected distance in-vivo is 250 μm with small frequency decay rates, while for in-vitro is 50μm with considerable frequency decay rates. Therefore, the developed model can be used for designing sensible light stimulation strategies in-vivo and opto-electronics systems.
Collapse
|
109
|
Bub G, Daniels MJ. Feasibility of Using Adjunctive Optogenetic Technologies in Cardiomyocyte Phenotyping - from the Single Cell to the Whole Heart. Curr Pharm Biotechnol 2020; 21:752-764. [PMID: 30961485 PMCID: PMC7527548 DOI: 10.2174/1389201020666190405182251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/21/2018] [Accepted: 03/20/2019] [Indexed: 12/21/2022]
Abstract
In 1791, Galvani established that electricity activated excitable cells. In the two centuries that followed, electrode stimulation of neuronal, skeletal and cardiac muscle became the adjunctive method of choice in experimental, electrophysiological, and clinical arenas. This approach underpins breakthrough technologies like implantable cardiac pacemakers that we currently take for granted. However, the contact dependence, and field stimulation that electrical depolarization delivers brings inherent limitations to the scope and experimental scale that can be achieved. Many of these were not exposed until reliable in vitro stem-cell derived experimental materials, with genotypes of interest, were produced in the numbers needed for multi-well screening platforms (for toxicity or efficacy studies) or the 2D or 3D tissue surrogates required to study propagation of depolarization within multicellular constructs that mimic clinically relevant arrhythmia in the heart or brain. Here the limitations of classical electrode stimulation are discussed. We describe how these are overcome by optogenetic tools which put electrically excitable cells under the control of light. We discuss how this enables studies in cardiac material from the single cell to the whole heart scale. We review the current commercial platforms that incorporate optogenetic stimulation strategies, and summarize the global literature to date on cardiac applications of optogenetics. We show that the advantages of optogenetic stimulation relevant to iPS-CM based screening include independence from contact, elimination of electrical stimulation artefacts in field potential measuring approaches such as the multi-electrode array, and the ability to print re-entrant patterns of depolarization at will on 2D cardiomyocyte monolayers.
Collapse
Affiliation(s)
| | - Matthew J. Daniels
- Address correspondence to this author at the Institute of Cardiovascular Sciences, University of Manchester, Core Technology Facility, 46 Grafton Street, Manchester, M13 9NT, UK; Tel: +441865234913; E-mails: ;
| |
Collapse
|
110
|
Wireless, battery-free, fully implantable multimodal and multisite pacemakers for applications in small animal models. Nat Commun 2019; 10:5742. [PMID: 31848334 PMCID: PMC6917818 DOI: 10.1038/s41467-019-13637-w] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 11/12/2019] [Indexed: 11/09/2022] Open
Abstract
Small animals support a wide range of pathological phenotypes and genotypes as versatile, affordable models for pathogenesis of cardiovascular diseases and for exploration of strategies in electrotherapy, gene therapy, and optogenetics. Pacing tools in such contexts are currently limited to tethered embodiments that constrain animal behaviors and experimental designs. Here, we introduce a highly miniaturized wireless energy-harvesting and digital communication electronics for thin, miniaturized pacing platforms weighing 110 mg with capabilities for subdermal implantation and tolerance to over 200,000 multiaxial cycles of strain without degradation in electrical or optical performance. Multimodal and multisite pacing in ex vivo and in vivo studies over many days demonstrate chronic stability and excellent biocompatibility. Optogenetic stimulation of cardiac cycles with in-animal control and induction of heart failure through chronic pacing serve as examples of modes of operation relevant to fundamental and applied cardiovascular research and biomedical technology. Pacing tools that support small animals and can serve as models for pathogenesis of cardiovascular diseases are currently not available. Here, the authors report a miniaturized wireless battery-free implantable multimodal and multisite pacemaker that provides unlimited stimulation to test subjects.
Collapse
|
111
|
Zgierski-Johnston CM, Ayub S, Fernández MC, Rog-Zielinska EA, Barz F, Paul O, Kohl P, Ruther P. Cardiac pacing using transmural multi-LED probes in channelrhodopsin-expressing mouse hearts. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 154:51-61. [PMID: 31738979 PMCID: PMC7322525 DOI: 10.1016/j.pbiomolbio.2019.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/07/2019] [Accepted: 11/13/2019] [Indexed: 02/03/2023]
Abstract
Optogenetics enables cell-type specific monitoring and actuation via light-activated proteins. In cardiac research, expressing light-activated depolarising ion channels in cardiomyocytes allows optical pacing and defibrillation. Previous studies largely relied on epicardial illumination. Light penetration through the myocardium is however problematic when moving to larger animals and humans. To overcome this limitation, we assessed the utility of an implantable multi light-emitting diode (LED) optical probe (IMLOP) for intramural pacing of mouse hearts expressing cardiac-specific channelrhodopsin-2 (ChR2). Here we demonstrated that IMLOP insertion needs approximately 20 mN of force, limiting possible damage from excessive loads applied during implantation. Histological sections confirmed the confined nature of tissue damage during acute use. The temperature change of the surrounding tissue was below 1 K during LED operation, rendering the probe safe for use in situ. This was confirmed in control experiments where no effect on cardiac action potential conduction was observed even when using stimulation parameters twenty-fold greater than required for pacing. In situ experiments on ChR2-expressing mouse hearts demonstrated that optical stimulation is possible with light intensities as low as 700 μW/mm2; although stable pacing requires higher intensities. When pacing with a single LED, rheobase and chronaxie values were 13.3 mW/mm2 ± 0.9 mW/mm2 and 3 ms ± 0.6 ms, respectively. When doubling the stimulated volume the rheobase decreased significantly (6.5 mW/mm2 ± 0.9 mW/mm2). We have demonstrated IMLOP-based intramural optical pacing of the heart. Probes cause locally constrained tissue damage in the acute setting and require low light intensities for pacing. Further development is necessary to assess effects of chronic implantation.
Collapse
Affiliation(s)
- C 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.
| | - S Ayub
- Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - M 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
| | - E A Rog-Zielinska
- 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
| | - F Barz
- Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - O Paul
- Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany; Cluster of Excellence BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| | - P Kohl
- 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
| | - P Ruther
- Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany; Cluster of Excellence BrainLinks-BrainTools, University of Freiburg, Freiburg, Germany
| |
Collapse
|
112
|
Liu K, Wang L. Optogenetics: Therapeutic spark in neuropathic pain. Bosn J Basic Med Sci 2019; 19:321-327. [PMID: 30995901 DOI: 10.17305/bjbms.2019.4114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/11/2019] [Indexed: 01/14/2023] Open
Abstract
Optogenetics is an emerging field, which uses light and molecular genetics to manipulate the activity of live cells by expressing light-sensitive proteins. With the discovery of bacteriorhodopsin, a light-sensitive bacterial protein, in 1971 Oesterhelt and Stoeckenius laid the pavement of optogenetics. However, the cross-integration of different disciplines is a little more than a decade old. The toolbox contains fluorescent sensors and optogenetic actuators that enable visualization of signaling events and manipulation of cellular activities, respectively. Neuropathic pain is pain caused either by damage or disease that affects the somatosensory system. The exact mechanism for neuropathic pain is not known, however proposed mechanisms include immune reactions, ion channel expressions, and inflammation. Current regimen for the disease provides about 50% relief for only 40-60% of patients. Recent in vivo and in vitro studies demonstrate the potential therapeutic applications of optogenetics by manipulating the activity of neurons. This review summarizes the basic concept, therapeutic applications for neuropathy, and potential of optogenetics to reach from bench to bedside in the near future.
Collapse
Affiliation(s)
- Kang Liu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China.
| | | |
Collapse
|
113
|
Keshmiri Neghab H, Goliaei B, Saboury AA, Esmaeeli Djavid G, Pornour M, Hong J, Grusch M. Modulation of cardiac optogenetics by vitamin A. Biofactors 2019; 45:983-990. [PMID: 31509323 DOI: 10.1002/biof.1564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 08/16/2019] [Indexed: 01/03/2023]
Abstract
Cardiac optogenetics is an emergent research area and refers to the delivery of light-activated proteins to excitable heart tissue and the subsequent use of light for controlling the electrical function with high spatial and temporal resolution. Channelrhodopsin-2 (ChR2) is a light-sensitive ion channel with the chromophore, all trans retinal, derived from vitamin A (all-trans-retinol; retinol). In this study, we explored whether exogenous vitamin A can be a limiting factor in the light responsiveness of cardiomyocytes-expressing ChR2. We showed that in cardiomyocytes virally transduced with ChR2 (H134R)-enhanced yellow fluorescent protein, vitamin A supplements lower than 10 μM significantly increased ChR2 expression. Adding 1 μM vitamin A changed light-induced transmembrane potential difference significantly, whereas 5 μM dramatically induced membrane depolarization and triggered intracellular calcium elevation. We concluded that vitamin A supplementation can modulate the efficiency of ChR2 and provide a complementary strategy for improving the performance of optogenetic tools.
Collapse
Affiliation(s)
| | - Bahram Goliaei
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali A Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Gholamreza Esmaeeli Djavid
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Majid Pornour
- Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Jun Hong
- School of Life Sciences, Henan University, Kaifeng, China
| | - Michael Grusch
- Department of Medicine I, Comprehensive Cancer Center Vienna, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
114
|
Living myofibroblast-silicon composites for probing electrical coupling in cardiac systems. Proc Natl Acad Sci U S A 2019; 116:22531-22539. [PMID: 31624124 DOI: 10.1073/pnas.1913651116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Traditional bioelectronics, primarily comprised of nonliving synthetic materials, lack cellular behaviors such as adaptability and motility. This shortcoming results in mechanically invasive devices and nonnatural signal transduction across cells and tissues. Moreover, resolving heterocellular electrical communication in vivo is extremely limited due to the invasiveness of traditional interconnected electrical probes. In this paper, we present a cell-silicon hybrid that integrates native cellular behavior (e.g., gap junction formation and biosignal processing) with nongenetically enabled photosensitivity. This hybrid configuration allows interconnect-free cellular modulation with subcellular spatial resolution for bioelectric studies. Specifically, we hybridize cardiac myofibroblasts with silicon nanowires and use these engineered hybrids to synchronize the electrical activity of cardiomyocytes, studying heterocellular bioelectric coupling in vitro. Thereafter, we inject the engineered myofibroblasts into heart tissues and show their ability to seamlessly integrate into contractile tissues in vivo. Finally, we apply local photostimulation with high cell specificity to tackle a long-standing debate regarding the existence of myofibroblast-cardiomyocyte electrical coupling in vivo.
Collapse
|
115
|
All AH, Zeng X, Teh DBL, Yi Z, Prasad A, Ishizuka T, Thakor N, Hiromu Y, Liu X. Expanding the Toolbox of Upconversion Nanoparticles for In Vivo Optogenetics and Neuromodulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803474. [PMID: 31432555 DOI: 10.1002/adma.201803474] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/12/2019] [Indexed: 06/10/2023]
Abstract
Optogenetics is an optical technique that exploits visible light for selective neuromodulation with spatio-temporal precision. Despite enormous effort, the effective stimulation of targeted neurons, which are located in deeper structures of the nervous system, by visible light, remains a technical challenge. Compared to visible light, near-infrared illumination offers a higher depth of tissue penetration owing to a lower degree of light attenuation. Herein, an overview of advances in developing new modalities for neural circuitry modulation utilizing upconversion-nanoparticle-mediated optogenetics is presented. These developments have led to minimally invasive optical stimulation and inhibition of neurons with substantially improved selectivity, sensitivity, and spatial resolution. The focus is to provide a comprehensive review of the mechanistic basis for evaluating upconversion parameters, which will be useful in designing, executing, and reporting optogenetic experiments.
Collapse
Affiliation(s)
- Angelo Homayoun All
- Department of Biomedical Engineering & Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Xiao Zeng
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Daniel Boon Loong Teh
- Department of Medicine & Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, 117456, Singapore
| | - Zhigao Yi
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Ankshita Prasad
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Toru Ishizuka
- Department of Integrative Life Sciences, Tohoku University Graduate School of Life Sciences, Sendai, 980-8577, Japan
| | - Nitish Thakor
- Department of Biomedical Engineering & Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Medicine & Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Yawo Hiromu
- Department of Integrative Life Sciences, Tohoku University Graduate School of Life Sciences, Sendai, 980-8577, Japan
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Center for Functional Materials, National University of Singapore Suzhou Research Institute, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
116
|
Jaimes R, McCullough D, Siegel B, Swift L, Hiebert J, Mclnerney D, Posnack NG. Lights, camera, path splitter: a new approach for truly simultaneous dual optical mapping of the heart with a single camera. BMC Biomed Eng 2019; 1. [PMID: 31768502 PMCID: PMC6876868 DOI: 10.1186/s42490-019-0024-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background Optical mapping of transmembrane voltage and intracellular calcium is a powerful tool for investigating cardiac physiology and pathophysiology. However, simultaneous dual mapping of two fluorescent probes remains technically challenging. We introduce a novel, easy-to-use approach that requires a path splitter, single camera and excitation light to simultaneously acquire voltage and calcium signals from whole heart preparations, which can be applied to other physiological models – including neurons and isolated cardiomyocytes. Results Complementary probes were selected that could be excited with a single wavelength light source. Langendorff-perfused hearts (rat, swine) were stained and imaged using a sCMOS camera outfitted with an optical path splitter to simultaneously acquire two emission fields at high spatial and temporal resolution. Voltage (RH237) and calcium (Rhod2) signals were acquired concurrently on a single sensor, resulting in two 384 × 256 images at 814 frames per second. At this frame rate, the signal-to-noise ratio was 47 (RH237) and 85 (Rhod2). Imaging experiments were performed on small rodent hearts, as well as larger pig hearts with sufficient optical signals. In separate experiments, each dye was used independently to assess crosstalk and demonstrate signal specificity. Additionally, the effect of ryanodine on myocardial calcium transients was validated – with no measurable effect on the amplitude of optical action potentials. To demonstrate spatial resolution, ventricular tachycardia was induced –resulting in the novel finding that spatially discordant calcium alternans can be present in different regions of the heart, even when electrical alternans remain concordant. The described system excels in providing a wide field of view and high spatiotemporal resolution for a variety of cardiac preparations. Conclusions We report the first multiparametric mapping system that simultaneously acquires calcium and voltage signals from cardiac preparations, using a path splitter, single camera and excitation light. This approach eliminates the need for multiple cameras, excitation light patterning or frame interleaving. These features can aid in the adoption of dual mapping technology by the broader cardiovascular research community, and decrease the barrier of entry into panoramic heart imaging, as it reduces the number of required cameras. Electronic supplementary material The online version of this article (10.1186/s42490-019-0024-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rafael Jaimes
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA.,Children's National Heart Institute: Children's National Health System, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Damon McCullough
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Bryan Siegel
- Children's National Heart Institute: Children's National Health System, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Luther Swift
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA.,Children's National Heart Institute: Children's National Health System, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - James Hiebert
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Daniel Mclnerney
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Nikki Gillum Posnack
- Sheikh Zayed Institute for Pediatric and Surgical Innovation: Children's National Health System, 6th floor, M7708, 111 Michigan Avenue NW, Washington, DC 20010, USA.,Children's National Heart Institute: Children's National Health System, 111 Michigan Avenue NW, Washington, DC 20010, USA.,Department of Pediatrics, Department of Pharmacology & Physiology, School of Medicine and Health Sciences: George Washington University, 2300 I Street NW, Washington, DC 20037, USA
| |
Collapse
|
117
|
Gruber A, Edri O, Gepstein L. Cardiac optogenetics: the next frontier. Europace 2019; 20:1910-1918. [PMID: 29315402 DOI: 10.1093/europace/eux371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
The emerging technology of optogenetics uses optical and genetic means to monitor and modulate the electrophysiological properties of excitable tissues. While transforming the field of neuroscience, the technology has recently gained popularity also in the cardiac arena. Here, we describe the basic principles of optogenetics, the available and evolving optogenetic tools, and the unique potential of this technology for basic and translational cardiac electrophysiology. Specifically, we discuss the ability to control (augment or suppress) the cardiac tissue's excitable properties using optogenetic actuators (microbial opsins), which are light-gated ion channels and pumps that can cause light-triggered membrane depolarization or hyperpolarization. We then focus on the potential clinical implications of this technology for the treatment of cardiac arrhythmias by describing recent efforts for developing optogenetic-based cardiac pacing, resynchronization, and defibrillation experimental strategies. Finally, the significant obstacles and challenges that need to be overcome before any future clinical translation can be expected are discussed.
Collapse
Affiliation(s)
- Amit Gruber
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Oded Edri
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Lior Gepstein
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel.,Cardiology Department of Rambam Health Care Campus, HaAliya HaShniya St 8, Haifa, Israel
| |
Collapse
|
118
|
Ferenczi EA, Tan X, Huang CLH. Principles of Optogenetic Methods and Their Application to Cardiac Experimental Systems. Front Physiol 2019; 10:1096. [PMID: 31572204 PMCID: PMC6749684 DOI: 10.3389/fphys.2019.01096] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/08/2019] [Indexed: 12/12/2022] Open
Abstract
Optogenetic techniques permit studies of excitable tissue through genetically expressed light-gated microbial channels or pumps permitting transmembrane ion movement. Light activation of these proteins modulates cellular excitability with millisecond precision. This review summarizes optogenetic approaches, using examples from neurobiological applications, and then explores their application in cardiac electrophysiology. We review the available opsins, including depolarizing and hyperpolarizing variants, as well as modulators of G-protein coupled intracellular signaling. We discuss the biophysical properties that determine the ability of microbial opsins to evoke reliable, precise stimulation or silencing of electrophysiological activity. We also review spectrally shifted variants offering possibilities for enhanced depth of tissue penetration, combinatorial stimulation for targeting different cell subpopulations, or all-optical read-in and read-out studies. Expression of the chosen optogenetic tool in the cardiac cell of interest then requires, at the single-cell level, introduction of opsin-encoding genes by viral transduction, or coupling "spark cells" to primary cardiomyocytes or a stem-cell derived counterpart. At the system-level, this requires construction of transgenic mice expressing ChR2 in their cardiomyocytes, or in vivo injection (myocardial or systemic) of adenoviral expression systems. Light delivery, by laser or LED, with widespread or multipoint illumination, although relatively straightforward in vitro may be technically challenged by cardiac motion and light-scattering in biological tissue. Physiological read outs from cardiac optogenetic stimulation include single cell patch clamp recordings, multi-unit microarray recordings from cell monolayers or slices, and electrical recordings from isolated Langendorff perfused hearts. Optical readouts of specific cellular events, including ion transients, voltage changes or activity in biochemical signaling cascades, using small detecting molecules or genetically encoded sensors now offer powerful opportunities for all-optical control and monitoring of cellular activity. Use of optogenetics has expanded in cardiac physiology, mainly using optically controlled depolarizing ion channels to control heart rate and for optogenetic defibrillation. ChR2-expressing cardiomyocytes show normal baseline and active excitable membrane and Ca2+ signaling properties and are sensitive even to ~1 ms light pulses. They have been employed in studies of the intrinsic cardiac adrenergic system and of cardiac arrhythmic properties.
Collapse
Affiliation(s)
- Emily A. Ferenczi
- Department of Neurology, Massachusetts General Hospital and Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Christopher L.-H. Huang
- Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
119
|
Richter C, Bruegmann T. No light without the dark: Perspectives and hindrances for translation of cardiac optogenetics. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 154:39-50. [PMID: 31515056 DOI: 10.1016/j.pbiomolbio.2019.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/18/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
Over the last decade, optogenetic stimulation of the heart and its translational potential for rhythm control attracted more and more interest. Optogenetics allows to stimulate cardiomyocytes expressing the light-gated cation channel Channelrhodopsin 2 (ChR2) with light and thus high spatio-temporal precision. Therefore this new approach can overcome the technical limitations of electrical stimulation. In regard of translational approaches, the prospect of pain-free stimulation, if ChR2 expression is restricted to cardiomyocytes, is especially intriguing and could be highly beneficial for cardioversion and defibrillation. However, there is no light without shadow and cardiac optogenetics has to surmount critical hurdles, namely "how" to inscribe light-sensitivity by expressing ChR2 in a native heart and how to avoid side effects such as possible immune responses against the gene transfer. Furthermore, implantable light devices have to be developed which ensure sufficient illumination in a highly contractile environment. Therefore this article reviews recent advantages in the field of cardiac optogenetics with a special focus on the hindrances for the potential translation of this new approach into clinics and provides an outlook how these have to be carefully investigated and could be solved step by step.
Collapse
Affiliation(s)
- Claudia Richter
- RG Biomedical Physics, Max Planck Institute for Dynamics & Self-Organization, Am Fassberg 17, 37077, Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center, Robert-Koch-Str. 42a, 37075, Goettingen, Germany; DZHK e.V. (German Center for Cardiovascular Research), Partner Site Goettingen, 37075, Goettingen, Germany.
| | - Tobias Bruegmann
- DZHK e.V. (German Center for Cardiovascular Research), Partner Site Goettingen, 37075, Goettingen, Germany; Institute for Cardiovascular Physiology, University Medical Center Goettingen, Humboldtallee 23, 37073, Goettingen, Germany.
| |
Collapse
|
120
|
Ye H, Fussenegger M. Optogenetic Medicine: Synthetic Therapeutic Solutions Precision-Guided by Light. Cold Spring Harb Perspect Med 2019; 9:a034371. [PMID: 30291146 PMCID: PMC6719591 DOI: 10.1101/cshperspect.a034371] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Gene- and cell-based therapies are well recognized as central pillars of next-generation medicine, but controllability remains a critical issue for clinical applications. In this context, optogenetics is opening up exciting new opportunities for precision-guided medicine by using illumination with light of appropriate intensity and wavelength as a trigger signal to achieve pinpoint spatiotemporal control of cellular activities, such as transgene expression. In this review, we highlight recent advances in optogenetics, focusing on devices for biomedical applications. We introduce the construction and applications of optogenetic-based biomedical tools to treat neurological diseases, diabetes, heart diseases, and cancer, as well as bioelectronic implants that combine light-interfaced electronic devices and optogenetic systems into portable personalized precision bioelectronic medical tools. Optogenetics-based technology promises the capability to achieve traceless, remotely controlled precision dosing of an enormous range of therapeutic outputs. Finally, we discuss the prospects for optogenetic medicine, as well as some emerging challenges.
Collapse
Affiliation(s)
- Haifeng Ye
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 200241 Shanghai, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, CH-4058 Basel, Switzerland
- University of Basel, Faculty of Science, CH-4058 Basel, Switzerland
| |
Collapse
|
121
|
Appiah C, Arndt C, Siemsen K, Heitmann A, Staubitz A, Selhuber-Unkel C. Living Materials Herald a New Era in Soft Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1807747. [PMID: 31267628 DOI: 10.1002/adma.201807747] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/07/2019] [Indexed: 05/22/2023]
Abstract
Living beings have an unsurpassed range of ways to manipulate objects and interact with them. They can make autonomous decisions and can heal themselves. So far, a conventional robot cannot mimic this complexity even remotely. Classical robots are often used to help with lifting and gripping and thus to alleviate the effects of menial tasks. Sensors can render robots responsive, and artificial intelligence aims at enabling autonomous responses. Inanimate soft robots are a step in this direction, but it will only be in combination with living systems that full complexity will be achievable. The field of biohybrid soft robotics provides entirely new concepts to address current challenges, for example the ability to self-heal, enable a soft touch, or to show situational versatility. Therefore, "living materials" are at the heart of this review. Similarly to biological taxonomy, there is a recent effort for taxonomy of biohybrid soft robotics. Here, an expansion is proposed to take into account not only function and origin of biohybrid soft robotic components, but also the materials. This materials taxonomy key demonstrates visually that materials science will drive the development of the field of soft biohybrid robotics.
Collapse
Affiliation(s)
- Clement Appiah
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Christine Arndt
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Katharina Siemsen
- Institute for Materials Science, University of Kiel, Kaiserstr. 2, D-24143, Kiel, Germany
| | - Anne Heitmann
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
| | - Anne Staubitz
- Institute for Organic and Analytical Chemistry, University of Bremen, Leobener Str. 7, D-28359, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bibliothekstraße 1, D-28359, Bremen, Germany
- Otto-Diels-Institute for Organic Chemistry, University of Kiel, Otto-Hahn-Platz 4, D-24118, Kiel, Germany
| | | |
Collapse
|
122
|
Stanley CE, Mauss AS, Borst A, Cooper RL. The Effects of Chloride Flux on Drosophila Heart Rate. Methods Protoc 2019; 2:mps2030073. [PMID: 31443492 PMCID: PMC6789470 DOI: 10.3390/mps2030073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022] Open
Abstract
Approaches are sought after to regulate ionotropic and chronotropic properties of the mammalian heart. Electrodes are commonly used for rapidly exciting cardiac tissue and resetting abnormal pacing. With the advent of optogenetics and the use of tissue-specific expression of light-activated channels, cardiac cells cannot only be excited but also inhibited with ion-selective conductance. As a proof of concept for the ability to slow down cardiac pacing, anion-conducting channelrhodopsins (GtACR1/2) and the anion pump halorhodopsin (eNpHR) were expressed in hearts of larval Drosophila and activated by light. Unlike body wall muscles in most animals, the equilibrium potential for Cl− is more positive as compared to the resting membrane potential in larval Drosophila. As a consequence, upon activating the two forms of GtACR1 and 2 with low light intensity the heart rate increased, likely due to depolarization and opening of voltage-gated Ca2+ channels. However, with very intense light activation the heart rate ceases, which may be due to Cl– shunting to the reversal potential for chloride. Activating eNpHR hyperpolarizes body wall and cardiac muscle in larval Drosophila and rapidly decreases heart rate. The decrease in heart rate is related to light intensity. Intense light activation of eNpHR stops the heart from beating, whereas lower intensities slowed the rate. Even with upregulation of the heart rate with serotonin, the pacing of the heart was slowed with light. Thus, regulation of the heart rate in Drosophila can be accomplished by activating anion-conducting channelrhodopsins using light. These approaches are demonstrated in a genetically amenable insect model.
Collapse
Affiliation(s)
- Catherine E Stanley
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA
| | - Alex S Mauss
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Alexander Borst
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Robin L Cooper
- Department of Biology, Center for Muscle Biology. University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
123
|
Dong R, Mu-u-min R, Reith AJM, O’Shea C, He S, Duan K, Kou K, Grassam-Rowe A, Tan X, Pavlovic D, Ou X, Lei M. A Protocol for Dual Calcium-Voltage Optical Mapping in Murine Sinoatrial Preparation With Optogenetic Pacing. Front Physiol 2019; 10:954. [PMID: 31456689 PMCID: PMC6698704 DOI: 10.3389/fphys.2019.00954] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 07/09/2019] [Indexed: 01/08/2023] Open
Abstract
Among the animal models for studying the molecular basis of atrial and sinoatrial node (SAN) biology and disease, the mouse is a widely used species due to its feasibility for genetic modifications in genes encoding ion channels or calcium handling and signaling proteins in the heart. It is therefore highly valuable to develop robust methodologies for studying SAN and atrial electrophysiological function in this species. Here, we describe a protocol for performing dual calcium-voltage optical mapping on mouse sinoatrial preparation (SAP), in combination with an optogenetic approach, for studying SAP membrane potential, intracellular Ca2+ transients, and pacemaker activity. The protocol includes the details for preparing the intact SAP, robust tissue dual-dye loading, light-programmed pacing, and high-resolution optical mapping. Our protocol provides an example of use of the combination of optogenetic and optical mapping techniques for investigating SAP membrane potential and intracellular Ca2+ transients and pacemaker activity with high temporal and spatial resolution in specific cardiac tissues. Thus, our protocol provides a useful tool for studying SAP physiology and pathophysiology in mice.
Collapse
Affiliation(s)
- Ruirui Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Razik Mu-u-min
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | | - Christopher O’Shea
- Institute for Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shicheng He
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Kaizhong Duan
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kun Kou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | | | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Davor Pavlovic
- Institute for Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
124
|
Ernst P, Xu N, Qu J, Chen H, Goldberg MS, Darley-Usmar V, Zhang JJ, O'Rourke B, Liu X, Zhou L. Precisely Control Mitochondria with Light to Manipulate Cell Fate Decision. Biophys J 2019; 117:631-645. [PMID: 31400914 DOI: 10.1016/j.bpj.2019.06.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/13/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
Abstract
Mitochondrial dysfunction has been implicated in many pathological conditions and diseases. The normal functioning of mitochondria relies on maintaining the inner mitochondrial membrane potential (also known as ΔΨm) that is essential for ATP synthesis, Ca2+ homeostasis, redox balance, and regulation of other key signaling pathways such as mitophagy and apoptosis. However, the detailed mechanisms by which ΔΨm regulates cellular function remain incompletely understood, partially because of the difficulty of manipulating ΔΨm with spatiotemporal resolution, reversibility, or cell type specificity. To address this need, we have developed a next generation optogenetic-based technique for controllable mitochondrial depolarization with light. We demonstrate successful targeting of the heterologous channelrhodopsin-2 fusion protein to the inner mitochondrial membrane and formation of functional cationic channels capable of light-induced selective ΔΨm depolarization and mitochondrial autophagy. Importantly, we for the first time, to our knowledge, show that optogenetic-mediated mitochondrial depolarization can be well controlled to differentially influence the fate of cells expressing mitochondrial channelrhodopsin-2; whereas sustained moderate light illumination induces substantial apoptotic cell death, transient mild light illumination elicits cytoprotection via mitochondrial preconditioning. Finally, we show that Parkin overexpression exacerbates, instead of ameliorating, mitochondrial depolarization-mediated cell death in HeLa cells. In summary, we provide evidence that the described mitochondrial-targeted optogenetics may have a broad application for studying the role of mitochondria in regulating cell function and fate decision.
Collapse
Affiliation(s)
- Patrick Ernst
- Departments of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama; Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ningning Xu
- Departments of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jing Qu
- Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Herbert Chen
- Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | - Jianyi J Zhang
- Departments of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | - Xiaoguang Liu
- Departments of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lufang Zhou
- Departments of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama; Medicine, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
125
|
Jiang C, Li HT, Zhou YM, Wang X, Wang L, Liu ZQ. Cardiac optogenetics: a novel approach to cardiovascular disease therapy. Europace 2019; 20:1741-1749. [PMID: 29253159 DOI: 10.1093/europace/eux345] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Optogenetics is a cell-type specific and high spatial-temporal resolution method that combines genetic encoding of light-sensitive proteins and optical manipulation techniques. Optogenetics technology provides a novel approach for research on cardiac arrhythmia treatment, including pacing, recovering the conduction system, and achieving cardiac resynchronization with precise and low-energy optical control. Photosensitive proteins, which usually act as ion channels, pumps, or receptors, are delivered to target cells, where they respond to light pulses of specific wavelengths, evoke transient flows of transmembrane ion currents, and induce signal transmission. With the development of gene technology, the in vivo efficiency of optogenetics in cardiology has been trialed, and in vitro experiments have been performed to test its potential in cardiac electrophysiology. Challenges for applying optogenetics in large animals and humans include the effectiveness, safety, and long-term expression of photosensitive proteins, unscattered and unattenuated exogenous light stimulation, and the need for implantable miniature light stimulators. Photosensitive proteins, genetic engineering technology, and light equipment are essential for experiments in cardiac optogenetics. Optogenetics may provide an alternative method for evaluating the mechanism of cardiac arrhythmias, testing hypotheses, and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Chan Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Hai Tao Li
- Department of Cardiology, Hainan General Hospital, Haikou, PR China
| | - Yong Ming Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China.,Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, PR China
| | - Zi Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.,Cardiovascular Research Institute, Wuhan University, Wuhan, PR China.,Hubei Key Laboratory of Cardiology, Wuhan, PR China
| |
Collapse
|
126
|
Sasse P, Funken M, Beiert T, Bruegmann T. Optogenetic Termination of Cardiac Arrhythmia: Mechanistic Enlightenment and Therapeutic Application? Front Physiol 2019; 10:675. [PMID: 31244670 PMCID: PMC6563676 DOI: 10.3389/fphys.2019.00675] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/13/2019] [Indexed: 01/08/2023] Open
Abstract
Optogenetic methods enable selective de- and hyperpolarization of cardiomyocytes expressing light-sensitive proteins within the myocardium. By using light, this technology provides very high spatial and temporal precision, which is in clear contrast to electrical stimulation. In addition, cardiomyocyte-specific expression would allow pain-free stimulation. In light of these intrinsic technical advantages, optogenetic methods provide an intriguing opportunity to understand and improve current strategies to terminate cardiac arrhythmia as well as for possible pain-free arrhythmia termination in patients in the future. In this review, we give a concise introduction to optogenetic stimulation of cardiomyocytes and the whole heart and summarize the recent progress on optogenetic defibrillation and cardioversion to terminate cardiac arrhythmia. Toward this aim, we specifically focus on the different mechanisms of optogenetic arrhythmia termination and how these might influence the prerequisites for success. Furthermore, we critically discuss the clinical perspectives and potential patient populations, which might benefit from optogenetic defibrillation devices.
Collapse
Affiliation(s)
- Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Maximilian Funken
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany.,Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thomas Beiert
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany.,Institute of Cardiovascular Physiology, University Medical Center, Georg August University Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| |
Collapse
|
127
|
Affiliation(s)
- Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2Q, 79110 Freiburg, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, Faculty of Medicine, University Heart Center, Medical Center-University of Freiburg, Elsässer Str. 2Q, 79110 Freiburg, Germany.,Institute for Physiology, Faculty of Medicine Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
128
|
Flexible and precise control of cardiac rhythm with blue light. Biochem Biophys Res Commun 2019; 514:759-764. [DOI: 10.1016/j.bbrc.2019.05.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/04/2019] [Indexed: 12/24/2022]
|
129
|
Gepstein L, Gruber A. Optogenetic Neuromodulation of the Heart. J Am Coll Cardiol 2019; 70:2791-2794. [PMID: 29191328 DOI: 10.1016/j.jacc.2017.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Lior Gepstein
- Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel; Cardiology Department, Rambam Health Care Campus, Haifa, Israel.
| | - Amit Gruber
- Sohnis Family Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
130
|
Zaglia T, Di Bona A, Mongillo M. A Light Wand to Untangle the Myocardial Cell Network. Methods Protoc 2019; 2:E34. [PMID: 31164614 PMCID: PMC6632158 DOI: 10.3390/mps2020034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/30/2022] Open
Abstract
The discovery of optogenetics has revolutionized research in neuroscience by providing the tools for noninvasive, cell-type selective modulation of membrane potential and cellular function in vitro and in vivo. Rhodopsin-based optogenetics has later been introduced in experimental cardiology studies and used as a tool to photoactivate cardiac contractions or to identify the sites, timing, and location most effective for defibrillating impulses to interrupt cardiac arrhythmias. The exploitation of cell-selectivity of optogenetics, and the generation of model organisms with myocardial cell type targeted expression of opsins has started to yield novel and sometimes unexpected notions on myocardial biology. This review summarizes the main results, the different uses, and the prospective developments of cardiac optogenetics.
Collapse
Affiliation(s)
- Tania Zaglia
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy.
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35122 Padova, Italy.
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy.
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy.
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy.
| | - Marco Mongillo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, via Giustiniani 2, 35128 Padova, Italy.
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35122 Padova, Italy.
- CNR Institute of Neuroscience, Viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
131
|
Funken M, Malan D, Sasse P, Bruegmann T. Optogenetic Hyperpolarization of Cardiomyocytes Terminates Ventricular Arrhythmia. Front Physiol 2019; 10:498. [PMID: 31105593 PMCID: PMC6491897 DOI: 10.3389/fphys.2019.00498] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/08/2019] [Indexed: 01/04/2023] Open
Abstract
Cardiac defibrillation to terminate lethal ventricular arrhythmia (VA) is currently performed by applying high energy electrical shocks. In cardiac tissue, electrical shocks induce simultaneously de- and hyperpolarized areas and only depolarized areas are considered to be responsible for VA termination. Because electrical shocks do not allow proper control over spatial extent and level of membrane potential changes, the effects of hyperpolarization have not been explored in the intact heart. In contrast, optogenetic methods allow cell type-selective induction of de- and hyperpolarization with unprecedented temporal and spatial control. To investigate effects of cardiomyocyte hyperpolarization on VA termination, we generated a mouse line with cardiomyocyte-specific expression of the light-driven proton pump ArchT. Isolated cardiomyocytes showed light-induced outward currents and hyperpolarization. Free-running VA were evoked by electrical stimulation of explanted hearts perfused with low K+ and the KATP channel opener Pinacidil. Optogenetic hyperpolarization was induced by epicardial illumination, which terminated VA with an average efficacy of ∼55%. This value was significantly higher compared to control hearts without illumination or ArchT expression (p = 0.0007). Intracellular recordings with sharp electrodes within the intact heart revealed hyperpolarization and faster action potential upstroke upon illumination, which should fasten conduction. However, conduction speed was lower during illumination suggesting enhanced electrical sink by hyperpolarization underlying VA termination. Thus, selective hyperpolarization in cardiomyocytes is able to terminate VA with a completely new mechanism of increased electrical sink. These novel insights could improve our mechanistic understanding and treatment strategies of VA termination.
Collapse
Affiliation(s)
- Maximilian Funken
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany.,Department of Internal Medicine II, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Daniela Malan
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany.,Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
132
|
Dwenger M, Kowalski WJ, Ye F, Yuan F, Tinney JP, Setozaki S, Nakane T, Masumoto H, Campbell P, Guido W, Keller BB. Chronic optical pacing conditioning of h-iPSC engineered cardiac tissues. J Tissue Eng 2019; 10:2041731419841748. [PMID: 31024681 PMCID: PMC6472158 DOI: 10.1177/2041731419841748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
The immaturity of human induced pluripotent stem cell derived engineered cardiac
tissues limits their ability to regenerate damaged myocardium and to serve as
robust in vitro models for human disease and drug toxicity
studies. Several chronic biomimetic conditioning protocols, including mechanical
stretch, perfusion, and/or electrical stimulation promote engineered cardiac
tissue maturation but have significant technical limitations. Non-contacting
chronic optical stimulation using heterologously expressed channelrhodopsin
light-gated ion channels, termed optogenetics, may be an advantageous
alternative to chronic invasive electrical stimulation for engineered cardiac
tissue conditioning. We designed proof-of-principle experiments to successfully
transfect human induced pluripotent stem cell derived engineered cardiac tissues
with a desensitization resistant, chimeric channelrhodopsin protein, and then
optically paced engineered cardiac tissues to accelerate maturation. We
transfected human induced pluripotent stem cell engineered cardiac tissues using
an adeno-associated virus packaged chimeric channelrhodopsin and then verified
optically paced by whole cell patch clamp. Engineered cardiac tissues were then
chronically optically paced above their intrinsic beat rates in
vitro from day 7 to 14. Chronically optically paced resulted in
improved engineered cardiac tissue electrophysiological properties and subtle
changes in the expression of some cardiac relevant genes, though active force
generation and histology were unchanged. These results validate the feasibility
of a novel chronically optically paced paradigm to explore non-invasive and
scalable optically paced–induced engineered cardiac tissue maturation
strategies.
Collapse
Affiliation(s)
- Marc Dwenger
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William J Kowalski
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA.,Laboratory of Stem Cell and Neurovascular Biology, Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Fei Ye
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Fangping Yuan
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Joseph P Tinney
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Shuji Setozaki
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Cardiovascular Surgery, Okamura Memorial Hospital, Shimizu, Japan
| | - Takeichiro Nakane
- 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 (CiRA), Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,RIKEN Center for Biosystems Dynamics Research (BDR), Wako, Japan
| | - Peter Campbell
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Bradley B Keller
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA.,Department of Pharmacology & Toxicology, School of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pediatrics, School of Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
133
|
Makowka P, Bruegmann T, Dusend V, Malan D, Beiert T, Hesse M, Fleischmann BK, Sasse P. Optogenetic stimulation of G s-signaling in the heart with high spatio-temporal precision. Nat Commun 2019; 10:1281. [PMID: 30894542 PMCID: PMC6426906 DOI: 10.1038/s41467-019-09322-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/25/2019] [Indexed: 01/08/2023] Open
Abstract
The standard technique for investigating adrenergic effects on heart function is perfusion with pharmaceutical agonists, which does not provide high temporal or spatial precision. Herein we demonstrate that the light sensitive Gs-protein coupled receptor JellyOp enables optogenetic stimulation of Gs-signaling in cardiomyocytes and the whole heart. Illumination of transgenic embryonic stem cell-derived cardiomyocytes or of the right atrium of mice expressing JellyOp elevates cAMP levels and instantaneously accelerates spontaneous beating rates similar to pharmacological β-adrenergic stimulation. Light application to the dorsal left atrium instead leads to supraventricular extrabeats, indicating adverse effects of localized Gs-signaling. In isolated ventricular cardiomyocytes from JellyOp mice, we find increased Ca2+ currents, fractional cell shortening and relaxation rates after illumination enabling the analysis of differential Gs-signaling with high temporal precision. Thus, JellyOp expression allows localized and time-restricted Gs stimulation and will provide mechanistic insights into different effects of site-specific, long-lasting and pulsatile Gs activation. Studying adrenergic signalling in the heart requires perfusion with receptor agonists, which lacks cell specificity and spatiotemporal control. Here the authors use the light sensitive G-coupled receptor JellyOp to optogenetically control Gs-signaling in cardiomyocytes and intact hearts with high spatiotemporal precision.
Collapse
Affiliation(s)
- Philipp Makowka
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127, Bonn, Germany.,Institute of Cardiovascular Physiology, University Medical Center, 37077, Göttingen, Germany
| | - Vanessa Dusend
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany.,Research Training Group 1873, University of Bonn, 53127, Bonn, Germany
| | - Daniela Malan
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Thomas Beiert
- Department of Internal Medicine II, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Michael Hesse
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, 53127, Bonn, Germany.
| |
Collapse
|
134
|
O’Shea C, Holmes AP, Winter J, Correia J, Ou X, Dong R, He S, Kirchhof P, Fabritz L, Rajpoot K, Pavlovic D. Cardiac Optogenetics and Optical Mapping - Overcoming Spectral Congestion in All-Optical Cardiac Electrophysiology. Front Physiol 2019; 10:182. [PMID: 30899227 PMCID: PMC6416196 DOI: 10.3389/fphys.2019.00182] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/14/2019] [Indexed: 12/30/2022] Open
Abstract
Optogenetic control of the heart is an emergent technology that offers unparalleled spatio-temporal control of cardiac dynamics via light-sensitive ion pumps and channels (opsins). This fast-evolving technique holds broad scope in both clinical and basic research setting. Combination of optogenetics with optical mapping of voltage or calcium fluorescent probes facilitates 'all-optical' electrophysiology, allowing precise optogenetic actuation of cardiac tissue with high spatio-temporal resolution imaging of action potential and calcium transient morphology and conduction patterns. In this review, we provide a synopsis of optogenetics and discuss in detail its use and compatibility with optical interrogation of cardiac electrophysiology. We briefly discuss the benefits of all-optical cardiac control and electrophysiological interrogation compared to traditional techniques, and describe mechanisms, unique features and limitations of optically induced cardiac control. In particular, we focus on state-of-the-art setup design, challenges in light delivery and filtering, and compatibility of opsins with fluorescent reporters used in optical mapping. The interaction of cardiac tissue with light, and physical and computational approaches to overcome the 'spectral congestion' that arises from the combination of optogenetics and optical mapping are discussed. Finally, we summarize recent preclinical work applications of combined cardiac optogenetics and optical mapping approach.
Collapse
Affiliation(s)
- Christopher O’Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
- EPSRC Centre for Doctoral Training in Physical Sciences for Health, School of Chemistry, University of Birmingham, Birmingham, United Kingdom
| | - Andrew P. Holmes
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - James Winter
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joao Correia
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ruirui Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Shicheng He
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease/Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, UHB NHS Trust, Birmingham, United Kingdom
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Cardiology, UHB NHS Trust, Birmingham, United Kingdom
| | - Kashif Rajpoot
- School of Computer Science, University of Birmingham, Birmingham, United Kingdom
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
135
|
Callaghan NI, Hadipour-Lakmehsari S, Lee SH, Gramolini AO, Simmons CA. Modeling cardiac complexity: Advancements in myocardial models and analytical techniques for physiological investigation and therapeutic development in vitro. APL Bioeng 2019; 3:011501. [PMID: 31069331 PMCID: PMC6481739 DOI: 10.1063/1.5055873] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023] Open
Abstract
Cardiomyopathies, heart failure, and arrhythmias or conduction blockages impact millions of patients worldwide and are associated with marked increases in sudden cardiac death, decline in the quality of life, and the induction of secondary pathologies. These pathologies stem from dysfunction in the contractile or conductive properties of the cardiomyocyte, which as a result is a focus of fundamental investigation, drug discovery and therapeutic development, and tissue engineering. All of these foci require in vitro myocardial models and experimental techniques to probe the physiological functions of the cardiomyocyte. In this review, we provide a detailed exploration of different cell models, disease modeling strategies, and tissue constructs used from basic to translational research. Furthermore, we highlight recent advancements in imaging, electrophysiology, metabolic measurements, and mechanical and contractile characterization modalities that are advancing our understanding of cardiomyocyte physiology. With this review, we aim to both provide a biological framework for engineers contributing to the field and demonstrate the technical basis and limitations underlying physiological measurement modalities for biologists attempting to take advantage of these state-of-the-art techniques.
Collapse
Affiliation(s)
| | | | | | | | - Craig A. Simmons
- Author to whom correspondence should be addressed: . Present address: Ted Rogers Centre for Heart
Research, 661 University Avenue, 14th Floor Toronto, Ontario M5G 1M1, Canada. Tel.:
416-946-0548. Fax: 416-978-7753
| |
Collapse
|
136
|
George SA, Efimov IR. Optocardiography: A Review of its Past, Present and Future. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019; 9:74-80. [PMID: 31803858 PMCID: PMC6892455 DOI: 10.1016/j.cobme.2019.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiac electrophysiology has progressed in great strides since the electrical activity of the heart was first discovered in 1842 and documented using electrocardiography. Optical imaging of cardiac electrophysiology, or optocardiography, has seen many advances in recent years including panoramic imaging of the heart, alternating transillumination to image transmural electrical activity, optogenetic models and customizable 3D printed optical mapping systems. Most of these techniques were adopted from other fields of study and refined for cardiac electrophysiology purposes. The future of this field could see similar adaptations of photoacoustic tomography, structured light technology and optical coherence tomography contributing to optocardiography.
Collapse
Affiliation(s)
- Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, DC
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC
| |
Collapse
|
137
|
Challis RC, Ravindra Kumar S, Chan KY, Challis C, Beadle K, Jang MJ, Kim HM, Rajendran PS, Tompkins JD, Shivkumar K, Deverman BE, Gradinaru V. Systemic AAV vectors for widespread and targeted gene delivery in rodents. Nat Protoc 2019; 14:379-414. [PMID: 30626963 DOI: 10.1038/s41596-018-0097-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We recently developed adeno-associated virus (AAV) capsids to facilitate efficient and noninvasive gene transfer to the central and peripheral nervous systems. However, a detailed protocol for generating and systemically delivering novel AAV variants was not previously available. In this protocol, we describe how to produce and intravenously administer AAVs to adult mice to specifically label and/or genetically manipulate cells in the nervous system and organs, including the heart. The procedure comprises three separate stages: AAV production, intravenous delivery, and evaluation of transgene expression. The protocol spans 8 d, excluding the time required to assess gene expression, and can be readily adopted by researchers with basic molecular biology, cell culture, and animal work experience. We provide guidelines for experimental design and choice of the capsid, cargo, and viral dose appropriate for the experimental aims. The procedures outlined here are adaptable to diverse biomedical applications, from anatomical and functional mapping to gene expression, silencing, and editing.
Collapse
Affiliation(s)
- Rosemary C Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ken Y Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Collin Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Keith Beadle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Min J Jang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hyun Min Kim
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Pradeep S Rajendran
- Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, University of California, Los Angeles, Los Angeles, CA, USA
| | - John D Tompkins
- Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kalyanam Shivkumar
- Cardiac Arrhythmia Center and Neurocardiology Research Center of Excellence, University of California, Los Angeles, Los Angeles, CA, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
138
|
Moreno A, Endicott K, Skancke M, Dwyer MK, Brennan J, Efimov IR, Trachiotis G, Mendelowitz D, Kay MW. Sudden Heart Rate Reduction Upon Optogenetic Release of Acetylcholine From Cardiac Parasympathetic Neurons in Perfused Hearts. Front Physiol 2019; 10:16. [PMID: 30745877 PMCID: PMC6360159 DOI: 10.3389/fphys.2019.00016] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/10/2019] [Indexed: 11/13/2022] Open
Abstract
The balance of sympathetic and parasympathetic tone provides exquisite control of heart rate and contractility and has also been shown to modulate coronary flow and inflammation. Understanding how autonomic balance is altered by cardiac disease is an active area of research, and developing new ways to control this balance provides insights into disease therapies. However, achieving acute neuron-specific stimulation of autonomic neurons can be difficult in experiments that measure the acute effects of nerve stimulation on the heart. Conventional electrical and pharmacological approaches can be spatially and temporally non-selective. Cell-specific expression of light-activated channels (channelrhodopsin, ChR2) is a powerful approach that enables control of the timing and distribution of cellular stimulation using light. We present such an optogenetic approach where parasympathetic cardiac neurons are selectively photoactivated at high temporal precision to initiate cholinergic-mediated slowing of heart rate. Mice were crossbred to express ChR2 in peripheral cholinergic neurons using Cre-Lox recombination driven by a choline acetyltransferase (ChAT) promoter. Hearts from adult mice were excised, perfused, and the epicardium was illuminated (peak 460–465 nm) to photoactivate ChR2. In one set of studies, hearts were illuminated using a large-field LED light source. In other studies, a micro LED was placed on the right atrium to selectively illuminate the junction of the superior vena cava (SVC) and right atrium. The ECG was acquired before, during, and after tissue illumination to measure changes in heart rate. Upon illumination, hearts exhibited sudden and dramatic reductions in heart rate with restoration of normal heart rate after cessation of illumination. Delays in atrioventricular conduction were also observed. Heart rate reductions at the highest irradiance levels were similar to heart rate reductions caused by application of bethanechol (10 μM) or acetylcholine (800 μM). Atropine (50 nM) completely blocked the effect of ChR2 photoactivation, confirming cholinergic mediation. Optogenetic activation of intrinsic parasympathetic neurons reduced heart rate in an immediate, dose-dependent fashion, resembling the slowing of sinus rate in response to acetylcholine. Our results demonstrate a new approach for controlling parasympathetic modulation of cardiac function by selectively activating the endogenous release of acetylcholine from intrinsic cardiac cholinergic neurons. Key Message: Optogenetic photoactivation of intrinsic cardiac neurons provides immediate, tissue-specific stimulation with minimal cross-reactivity. Our results demonstrate that selective expression of channelrhodopsin within cardiac cholinergic neurons enables photoactivated release of acetylcholine, thereby instantaneously slowing sinus rate and altering atrioventricular conduction. This provides for in-depth examination of the endogenous interplay between cardiac autonomic neurons and the functional outcomes of downstream post-synaptic receptor activation.
Collapse
Affiliation(s)
- Angel Moreno
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Kendal Endicott
- Division of Cardiothoracic Surgery, Veterans Affairs Medical Center, Washington, DC, United States
| | - Matthew Skancke
- Division of Cardiothoracic Surgery, Veterans Affairs Medical Center, Washington, DC, United States
| | - Mary Kate Dwyer
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| | - Jaclyn Brennan
- 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
| | - Gregory Trachiotis
- Division of Cardiothoracic Surgery, Veterans Affairs Medical Center, Washington, DC, United States
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, United States
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, DC, United States
| |
Collapse
|
139
|
A Software Architecture to Mimic a Ventricular Tachycardia in Intact Murine Hearts by Means of an All-Optical Platform. Methods Protoc 2019; 2:mps2010007. [PMID: 31164591 PMCID: PMC6481051 DOI: 10.3390/mps2010007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/29/2018] [Accepted: 01/04/2019] [Indexed: 11/17/2022] Open
Abstract
Optogenetics is an emerging method that uses light to manipulate electrical activity in excitable cells exploiting the interaction between light and light-sensitive depolarizing ion channels, such as channelrhodopsin-2 (ChR2). Initially used in the neuroscience, it has been adopted in cardiac research where the expression of ChR2 in cardiac preparations allows optical pacing, resynchronization and defibrillation. Recently, optogenetics has been leveraged to manipulate cardiac electrical activity in the intact heart in real-time. This new approach was applied to simulate a re-entrant circuit across the ventricle. In this technical note, we describe the development and the implementation of a new software package for real-time optogenetic intervention. The package consists of a single LabVIEW program that simultaneously captures images at very high frame rates and delivers precisely timed optogenetic stimuli based on the content of the images. The software implementation guarantees closed-loop optical manipulation at high temporal resolution by processing the raw data in workstation memory. We demonstrate that this strategy allows the simulation of a ventricular tachycardia with high stability and with a negligible loss of data with a temporal resolution of up to 1 ms.
Collapse
|
140
|
Kopton RA, Baillie JS, Rafferty SA, Moss R, Zgierski-Johnston CM, Prykhozhij SV, Stoyek MR, Smith FM, Kohl P, Quinn TA, Schneider-Warme F. Cardiac Electrophysiological Effects of Light-Activated Chloride Channels. Front Physiol 2018; 9:1806. [PMID: 30618818 PMCID: PMC6304430 DOI: 10.3389/fphys.2018.01806] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/30/2018] [Indexed: 12/17/2022] Open
Abstract
During the last decade, optogenetics has emerged as a paradigm-shifting technique to monitor and steer the behavior of specific cell types in excitable tissues, including the heart. Activation of cation-conducting channelrhodopsins (ChR) leads to membrane depolarization, allowing one to effectively trigger action potentials (AP) in cardiomyocytes. In contrast, the quest for optogenetic tools for hyperpolarization-induced inhibition of AP generation has remained challenging. The green-light activated ChR from Guillardia theta (GtACR1) mediates Cl--driven photocurrents that have been shown to silence AP generation in different types of neurons. It has been suggested, therefore, to be a suitable tool for inhibition of cardiomyocyte activity. Using single-cell electrophysiological recordings and contraction tracking, as well as intracellular microelectrode recordings and in vivo optical recordings of whole hearts, we find that GtACR1 activation by prolonged illumination arrests cardiac cells in a depolarized state, thus inhibiting re-excitation. In line with this, GtACR1 activation by transient light pulses elicits AP in rabbit isolated cardiomyocytes and in spontaneously beating intact hearts of zebrafish. Our results show that GtACR1 inhibition of AP generation is caused by cell depolarization. While this does not address the need for optogenetic silencing through physiological means (i.e., hyperpolarization), GtACR1 is a potentially attractive tool for activating cardiomyocytes by transient light-induced depolarization.
Collapse
Affiliation(s)
- Ramona A Kopton
- Institute for Experimental Cardiovascular Medicine, University Heart Centre 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
| | - Jonathan S Baillie
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Sara A Rafferty
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Robin Moss
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Callum M Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | | | - Matthew R Stoyek
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada
| | - Frank M Smith
- Department of Medical Neuroscience, Dalhousie University Halifax, NS, Canada
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University Halifax, NS, Canada.,School of Biomedical Engineering, Dalhousie University Halifax, NS, Canada
| | - Franziska Schneider-Warme
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg-Bad Krozingen Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine University of Freiburg, Freiburg, Germany
| |
Collapse
|
141
|
Quiñonez Uribe RA, Luther S, Diaz-Maue L, Richter C. Energy-Reduced Arrhythmia Termination Using Global Photostimulation in Optogenetic Murine Hearts. Front Physiol 2018; 9:1651. [PMID: 30542292 PMCID: PMC6277892 DOI: 10.3389/fphys.2018.01651] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/02/2018] [Indexed: 02/01/2023] Open
Abstract
Complex spatiotemporal non-linearity as observed during cardiac arrhythmia strongly correlates with vortex-like excitation wavelengths and tissue characteristics. Therefore, the control of arrhythmic patterns requires fundamental understanding of dependencies between onset and perpetuation of arrhythmia and substrate instabilities. Available treatments, such as drug application or high-energy electrical shocks, are discussed for potential side effects resulting in prognosis worsening due to the lack of specificity and spatiotemporal precision. In contrast, cardiac optogenetics relies on light sensitive ion channels stimulated to trigger excitation of cardiomyocytes solely making use of the inner cell mechanisms. This enables low-energy, non-damaging optical control of cardiac excitation with high resolution. Recently, the capability of optogenetic cardioversion was shown in Channelrhodopsin-2 (ChR2) transgenic mice. But these studies used mainly structured and local illumination for cardiac stimulation. In addition, since optogenetic and electrical stimulus work on different principles to control the electrical activity of cardiac tissue, a better understanding of the phenomena behind optogenetic cardioversion is still needed. The present study aims to investigate global illumination with regard to parameter characterization and its potential for cardioversion. Our results show that by tuning the light intensity without exceeding 1.10 mW mm-2, a single pulse in the range of 10–1,000 ms is sufficient to reliably reset the heart into sinus rhythm. The combination of our panoramic low-intensity photostimulation with optical mapping techniques visualized wave collision resulting in annihilation as well as propagation perturbations as mechanisms leading to optogenetic cardioversion, which seem to base on other processes than electrical defibrillation. This study contributes to the understanding of the roles played by epicardial illumination, pulse duration and light intensity in optogenetic cardioversion, which are the main variables influencing cardiac optogenetic control, highlighting the advantages and insights of global stimulation. Therefore, the presented results can be modules in the design of novel illumination technologies with specific energy requirements on the way toward tissue-protective defibrillation techniques.
Collapse
Affiliation(s)
- Raúl A Quiñonez Uribe
- RG Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Stefan Luther
- RG Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Institute for Nonlinear Dynamics, Georg-August University, Göttingen, Germany.,Department of Pharmacology and Toxicology, University Medical Center, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partner Site Göttingen, Göttingen, Germany
| | - Laura Diaz-Maue
- RG Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Claudia Richter
- RG Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partner Site Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center, Göttingen, Germany
| |
Collapse
|
142
|
Majumder R, Feola I, Teplenin AS, de Vries AA, Panfilov AV, Pijnappels DA. Optogenetics enables real-time spatiotemporal control over spiral wave dynamics in an excitable cardiac system. eLife 2018; 7:41076. [PMID: 30260316 PMCID: PMC6195347 DOI: 10.7554/elife.41076] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/14/2018] [Indexed: 11/19/2022] Open
Abstract
Propagation of non-linear waves is key to the functioning of diverse biological systems. Such waves can organize into spirals, rotating around a core, whose properties determine the overall wave dynamics. Theoretically, manipulation of a spiral wave core should lead to full spatiotemporal control over its dynamics. However, this theory lacks supportive evidence (even at a conceptual level), making it thus a long-standing hypothesis. Here, we propose a new phenomenological concept that involves artificially dragging spiral waves by their cores, to prove the aforementioned hypothesis in silico, with subsequent in vitro validation in optogenetically modified monolayers of rat atrial cardiomyocytes. We thereby connect previously established, but unrelated concepts of spiral wave attraction, anchoring and unpinning to demonstrate that core manipulation, through controlled displacement of heterogeneities in excitable media, allows forced movement of spiral waves along pre-defined trajectories. Consequently, we impose real-time spatiotemporal control over spiral wave dynamics in a biological system. From a spinning galaxy to a swarm of honeybees, rotating spirals are widespread in nature. Even within the muscles of the heart, waves of electrical activity sometimes rotate spirally, leading to irregular heart rhythms or arrhythmia – a condition that can be fatal. Irrespective of where they occur, spiral waves organize around a center or core with different biophysical properties compared to the rest of the medium. The properties of the core determine the overall dynamics of the spiral. This means that, theoretically, it should be possibly to completely control a spiral wave just by manipulating its core. Now, Majumder, Feola et al. have tested this long-standing hypothesis using a combination of computer modeling and experiments with single layers of rat heart cells grown in a laboratory. First, the heart cells were genetically modified so that their electrical properties could be altered with light; in other words, the cells were put under optical control. Next, by using of a narrow beam of light, Majumder, Feola et al. precisely controlled the electrical properties of a small number of cells, which then attracted and supported a rotating spiral wave by acting as its new core. Moving the light beam allowed the core of the spiral wave to be shifted too, meaning the spiral wave could now be steered along any desired path in the cell layer. Majumder, Feola et al. hope that these underlying principles may one day provide the basis of new treatments for irregular heartbeats that are more effective and less damaging to the heart than existing options. Yet first, more work is needed to translate these findings from single layers of cells to actual hearts.
Collapse
Affiliation(s)
- Rupamanjari Majumder
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Iolanda Feola
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexander S Teplenin
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Antoine Af de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexander V Panfilov
- Department of Physics and Astronomy, Gent University, Gent, Belgium.,Laboratory of Computational Biology and Medicine, Ural Federal University, Ekaterinburg, Russia
| | - Daniel A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
143
|
Ma G, Liu J, Nguyen NT, Zhou Y, Wang Y. Let there be light: a bright future for Ca 2+ signaling. Sci Bull (Beijing) 2018; 63:1029-1031. [PMID: 36755452 DOI: 10.1016/j.scib.2018.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Guolin Ma
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Jindou Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Nhung T Nguyen
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Yubin Zhou
- Center for Translational Cancer Research, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX 77030, USA.
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
144
|
Scardigli M, Müllenbroich C, Margoni E, Cannazzaro S, Crocini C, Ferrantini C, Coppini R, Yan P, Loew LM, Campione M, Bocchi L, Giulietti D, Cerbai E, Poggesi C, Bub G, Pavone FS, Sacconi L. Real-time optical manipulation of cardiac conduction in intact hearts. J Physiol 2018; 596:3841-3858. [PMID: 29989169 PMCID: PMC6117584 DOI: 10.1113/jp276283] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/05/2018] [Indexed: 11/28/2022] Open
Abstract
Key points Although optogenetics has clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies lack the capability to react acutely to ongoing cardiac wave dynamics. Here, we developed an all‐optical platform to monitor and control electrical activity in real‐time. The methodology was applied to restore normal electrical activity after atrioventricular block and to manipulate the intraventricular propagation of the electrical wavefront. The closed‐loop approach was also applied to simulate a re‐entrant circuit across the ventricle. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time all‐optical stimulation can control cardiac rhythm in normal and abnormal conditions.
Abstract Optogenetics has provided new insights in cardiovascular research, leading to new methods for cardiac pacing, resynchronization therapy and cardioversion. Although these interventions have clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies do not take into account cardiac wave dynamics in real time. Here, we developed an all‐optical platform complemented by integrated, newly developed software to monitor and control electrical activity in intact mouse hearts. The system combined a wide‐field mesoscope with a digital projector for optogenetic activation. Cardiac functionality could be manipulated either in free‐run mode with submillisecond temporal resolution or in a closed‐loop fashion: a tailored hardware and software platform allowed real‐time intervention capable of reacting within 2 ms. The methodology was applied to restore normal electrical activity after atrioventricular block, by triggering the ventricle in response to optically mapped atrial activity with appropriate timing. Real‐time intraventricular manipulation of the propagating electrical wavefront was also demonstrated, opening the prospect for real‐time resynchronization therapy and cardiac defibrillation. Furthermore, the closed‐loop approach was applied to simulate a re‐entrant circuit across the ventricle demonstrating the capability of our system to manipulate heart conduction with high versatility even in arrhythmogenic conditions. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time optically based stimulation can control cardiac rhythm in normal and abnormal conditions, promising a new approach for the investigation of the (patho)physiology of the heart. Although optogenetics has clearly demonstrated the feasibility of cardiac manipulation, current optical stimulation strategies lack the capability to react acutely to ongoing cardiac wave dynamics. Here, we developed an all‐optical platform to monitor and control electrical activity in real‐time. The methodology was applied to restore normal electrical activity after atrioventricular block and to manipulate the intraventricular propagation of the electrical wavefront. The closed‐loop approach was also applied to simulate a re‐entrant circuit across the ventricle. The development of this innovative optical methodology provides the first proof‐of‐concept that a real‐time all‐optical stimulation can control cardiac rhythm in normal and abnormal conditions.
Collapse
Affiliation(s)
- M Scardigli
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Müllenbroich
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - E Margoni
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,Department of Physics, University of Pisa, Pisa, 56127, Italy
| | - S Cannazzaro
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Crocini
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| | - C Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - R Coppini
- Division of Pharmacology, Department 'NeuroFarBa', University of Florence, Florence, 50139, Italy
| | - P Yan
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - L M Loew
- R. D. Berlin Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - M Campione
- Neuroscience Institute, National Research Council, Padova, 35121, Italy.,Department of Biomedical Sciences, Univercity ot Padua, Padua, 35121, Italy
| | - L Bocchi
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,Department of Information Engineering, University of Florence, Via S. Marta 3, Florence, 50139, Italy
| | - D Giulietti
- National Institute of Optics, National Research Council, Florence, 50125, Italy.,Department of Physics, University of Pisa, Pisa, 56127, Italy
| | - E Cerbai
- Division of Pharmacology, Department 'NeuroFarBa', University of Florence, Florence, 50139, Italy
| | - C Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, University of Florence, Florence, 50134, Italy
| | - G Bub
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - F S Pavone
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy.,Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, 50019, Italy
| | - L Sacconi
- European Laboratory for Non-Linear Spectroscopy, Florence, 50019, Italy.,National Institute of Optics, National Research Council, Florence, 50125, Italy
| |
Collapse
|
145
|
Fehrentz T, Huber FME, Hartrampf N, Bruegmann T, Frank JA, Fine NHF, Malan D, Danzl JG, Tikhonov DB, Sumser M, Sasse P, Hodson DJ, Zhorov BS, Klöcker N, Trauner D. Optical control of L-type Ca 2+ channels using a diltiazem photoswitch. Nat Chem Biol 2018; 14:764-767. [PMID: 30013061 DOI: 10.1038/s41589-018-0090-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/21/2018] [Indexed: 01/21/2023]
Abstract
L-type Ca2+ channels (LTCCs) play a crucial role in excitation-contraction coupling and release of hormones from secretory cells. They are targets of antihypertensive and antiarrhythmic drugs such as diltiazem. Here, we present a photoswitchable diltiazem, FHU-779, which can be used to reversibly block endogenous LTCCs by light. FHU-779 is as potent as diltiazem and can be used to place pancreatic β-cell function and cardiac activity under optical control.
Collapse
Affiliation(s)
- Timm Fehrentz
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany.
| | - Florian M E Huber
- Department of Chemistry, University of Munich and Center for Integrated Protein Science (CIPSM), Munich, Germany.,Roche Diagnostics GmbH, DXRERA, Penzberg, Germany
| | - Nina Hartrampf
- Department of Chemistry, University of Munich and Center for Integrated Protein Science (CIPSM), Munich, Germany
| | - Tobias Bruegmann
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - James A Frank
- Department of Chemistry, University of Munich and Center for Integrated Protein Science (CIPSM), Munich, Germany.,Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas H F Fine
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Daniela Malan
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - Johann G Danzl
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Denis B Tikhonov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Martin Sumser
- Department of Chemistry, University of Munich and Center for Integrated Protein Science (CIPSM), Munich, Germany
| | - Philipp Sasse
- Institute of Physiology I, Medical Faculty, University of Bonn, Bonn, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Boris S Zhorov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia.,Institute of Molecular Biology and Genetics, Almazov Federal Heart, Blood and Endocrinology Centre, St. Petersburg, Russia.,Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Nikolaj Klöcker
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany.
| | - Dirk Trauner
- Department of Chemistry, University of Munich and Center for Integrated Protein Science (CIPSM), Munich, Germany. .,Department of Chemistry and Neuroscience Institute, New York University, New York, NY, USA.
| |
Collapse
|
146
|
Chen S, Li R, Li X, Xie J. Electrospinning: An enabling nanotechnology platform for drug delivery and regenerative medicine. Adv Drug Deliv Rev 2018; 132:188-213. [PMID: 29729295 DOI: 10.1016/j.addr.2018.05.001] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/03/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
Electrospinning provides an enabling nanotechnology platform for generating a rich variety of novel structured materials in many biomedical applications including drug delivery, biosensing, tissue engineering, and regenerative medicine. In this review article, we begin with a thorough discussion on the method of producing 1D, 2D, and 3D electrospun nanofiber materials. In particular, we emphasize on how the 3D printing technology can contribute to the improvement of traditional electrospinning technology for the fabrication of 3D electrospun nanofiber materials as drug delivery devices/implants, scaffolds or living tissue constructs. We then highlight several notable examples of electrospun nanofiber materials in specific biomedical applications including cancer therapy, guiding cellular responses, engineering in vitro 3D tissue models, and tissue regeneration. Finally, we finish with conclusions and future perspectives of electrospun nanofiber materials for drug delivery and regenerative medicine.
Collapse
|
147
|
Prando V, Da Broi F, Franzoso M, Plazzo AP, Pianca N, Francolini M, Basso C, Kay MW, Zaglia T, Mongillo M. Dynamics of neuroeffector coupling at cardiac sympathetic synapses. J Physiol 2018; 596:2055-2075. [PMID: 29524231 PMCID: PMC5983210 DOI: 10.1113/jp275693] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/28/2018] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The present study demonstrates, by in vitro and in vivo analyses, the novel concept that signal transmission between sympathetic neurons and the heart, underlying the physiological regulation of cardiac function, operates in a quasi-synaptic fashion. This is a result of the direct coupling between neurotransmitter releasing sites and effector cardiomyocyte membranes. ABSTRACT Cardiac sympathetic neurons (SNs) finely tune the rate and strength of heart contractions to match blood demand, both at rest and during acute stress, through the release of noradrenaline (NE). Junctional sites at the interface between the two cell types have been observed, although whether direct neurocardiac coupling has a role in heart physiology has not been clearly demonstrated to date. We investigated the dynamics of SN/cardiomyocyte intercellular signalling, both by fluorescence resonance energy transfer-based imaging of cAMP in co-cultures, as a readout of cardiac β-adrenergic receptor activation, and in vivo, using optogenetics in transgenic mice with SN-specific expression of Channelrhodopsin-2. We demonstrate that SNs and cardiomyocytes interact at specific sites in the human and rodent heart, as well as in co-cultures. Accordingly, neuronal activation elicited intracellular cAMP increases only in directly contacted myocytes and cell-cell coupling utilized a junctional extracellular signalling domain with an elevated NE concentration. In the living mouse, optogenetic activation of cardiac SNs innervating the sino-atrial node resulted in an instantaneous chronotropic effect, which shortened the heartbeat interval with single beat precision. Remarkably, inhibition of the optogenetically elicited chronotropic responses required a high dose of propranolol (20-50 mg kg-1 ), suggesting that sympathetic neurotransmission in the heart occurs at a locally elevated NE concentration. Our in vitro and in vivo data suggest that the control of cardiac function by SNs occurs via direct intercellular coupling as a result of the establishment of a specific junctional site.
Collapse
Affiliation(s)
- Valentina Prando
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Mauro Franzoso
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Nicola Pianca
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | | | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular SceincesUniversity of PadovaPadovaItaly
| | - Matthew W. Kay
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDCUSA
| | - Tania Zaglia
- Venetian Institute of Molecular MedicinePadovaItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Department of Cardiac, Thoracic and Vascular SceincesUniversity of PadovaPadovaItaly
| | - Marco Mongillo
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- University of MilanoMilanoItaly
- CNR Institute of NeurosciencePadovaItaly
| |
Collapse
|
148
|
Rorsman NJG, Ta CM, Garnett H, Swietach P, Tammaro P. Defining the ionic mechanisms of optogenetic control of vascular tone by channelrhodopsin-2. Br J Pharmacol 2018; 175:2028-2045. [PMID: 29486056 PMCID: PMC5979753 DOI: 10.1111/bph.14183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/16/2018] [Accepted: 02/14/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Optogenetic control of electromechanical coupling in vascular smooth muscle cells (VSMCs) is emerging as a powerful research tool with potential applications in drug discovery and therapeutics. However, the precise ionic mechanisms involved in this control remain unclear. EXPERIMENTAL APPROACH Cell imaging, patch-clamp electrophysiology and muscle tension recordings were used to define these mechanisms over a wide range of light stimulations. KEY RESULTS Transgenic mice expressing a channelrhodopsin-2 variant [ChR2(H134R)] selectively in VSMCs were generated. Isolated VSMCs obtained from these mice demonstrated blue light-induced depolarizing whole-cell currents. Fine control of artery tone was attained by varying the intensity of the light stimulus. This arterial response was sufficient to overcome the endogenous, melanopsin-mediated, light-evoked, arterial relaxation observed in the presence of contractile agonists. Ca2+ entry through voltage-gated Ca2+ channels, and opening of plasmalemmal depolarizing channels (TMEM16A and TRPM) and intracellular IP3 receptors were involved in the ChR2(H134R)-dependent arterial response to blue light at intensities lower than ~0.1 mW·mm-2 . Light stimuli of greater intensity evoked a significant Ca2+ influx directly through ChR2(H134R) and produced marked intracellular alkalinization of VSMCs. CONCLUSIONS AND IMPLICATIONS We identified the range of light intensity allowing optical control of arterial tone, primarily by means of endogenous channels and without substantial alteration to intracellular pH. Within this range, mice expressing ChR2(H134R) in VSMCs are a powerful experimental model for achieving accurate and tuneable optical voltage-clamp of VSMCs and finely graded control of arterial tone, offering new approaches to the discovery of vasorelaxant drugs.
Collapse
Affiliation(s)
- Nils J G Rorsman
- Department of PharmacologyUniversity of OxfordOxfordUK
- OXION Initiative in Ion Channels and DiseaseUniversity of OxfordOxfordUK
| | - Chau M Ta
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | - Pawel Swietach
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordOxfordUK
| | - Paolo Tammaro
- Department of PharmacologyUniversity of OxfordOxfordUK
- OXION Initiative in Ion Channels and DiseaseUniversity of OxfordOxfordUK
| |
Collapse
|
149
|
Broyles CN, Robinson P, Daniels MJ. Fluorescent, Bioluminescent, and Optogenetic Approaches to Study Excitable Physiology in the Single Cardiomyocyte. Cells 2018; 7:cells7060051. [PMID: 29857560 PMCID: PMC6028913 DOI: 10.3390/cells7060051] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
This review briefly summarizes the single cell application of classical chemical dyes used to visualize cardiomyocyte physiology and their undesirable toxicities which have the potential to confound experimental observations. We will discuss, in detail, the more recent iterative development of fluorescent and bioluminescent protein-based indicators and their emerging application to cardiomyocytes. We will discuss the integration of optical control strategies (optogenetics) to augment the standard imaging approach. This will be done in the context of potential applications, and barriers, of these technologies to disease modelling, drug toxicity, and drug discovery efforts at the single-cell scale.
Collapse
Affiliation(s)
- Connor N Broyles
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
| | - Paul Robinson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
| | - Matthew J Daniels
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK.
- BHF Centre of Research Excellence, University of Oxford, Oxford OX3 9DU, UK.
- Department of Cardiology, Oxford University NHS Hospitals Trust, Oxford OX3 9DU, UK.
- BHF Centre of Regenerative Medicine, University of Oxford, Oxford OX3 9DU, UK.
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Mihogaoka 8-1, Ibaraki, 567-0047 Osaka, Japan.
| |
Collapse
|
150
|
Thomas K, Goudy J, Henley T, Bressan M. Optical Electrophysiology in the Developing Heart. J Cardiovasc Dev Dis 2018; 5:E28. [PMID: 29751595 PMCID: PMC6023508 DOI: 10.3390/jcdd5020028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
The heart is the first organ system to form in the embryo. Over the course of development, cardiomyocytes with differing morphogenetic, molecular, and physiological characteristics are specified and differentiate and integrate with one another to assemble a coordinated electromechanical pumping system that can function independently of any external stimulus. As congenital malformation of the heart presents the leading class of birth defects seen in humans, the molecular genetics of heart development have garnered much attention over the last half century. However, understanding how genetic perturbations manifest at the level of the individual cell function remains challenging to investigate. Some of the barriers that have limited our capacity to construct high-resolution, comprehensive models of cardiac physiological maturation are rapidly being removed by advancements in the reagents and instrumentation available for high-speed live imaging. In this review, we briefly introduce the history of imaging approaches for assessing cardiac development, describe some of the reagents and tools required to perform live imaging in the developing heart, and discuss how the combination of modern imaging modalities and physiological probes can be used to scale from subcellular to whole-organ analysis. Through these types of imaging approaches, critical insights into the processes of cardiac physiological development can be directly examined in real-time. Moving forward, the synthesis of modern molecular biology and imaging approaches will open novel avenues to investigate the mechanisms of cardiomyocyte maturation, providing insight into the etiology of congenital heart defects, as well as serving to direct approaches for designing stem-cell or regenerative medicine protocols for clinical application.
Collapse
Affiliation(s)
- Kandace Thomas
- Department of Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Julie Goudy
- Department of Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Trevor Henley
- Department of Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Michael Bressan
- Department of Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|