Modulating cardiac physiology in engineered heart tissue with the bidirectional optogenetic tool BiPOLES

Standard

Modulating cardiac physiology in engineered heart tissue with the bidirectional optogenetic tool BiPOLES. / Schwarzová, Barbora; Stüdemann, Tim; Sönmez, Muhammed; Rössinger, Judith; Pan, Bangfen; Eschenhagen, Thomas; Stenzig, Justus; Wiegert, J Simon; Christ, Torsten; Weinberger, Florian.

In: PFLUG ARCH EUR J PHY, Vol. 475, No. 12, 12.2023, p. 1463-1477.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

APA

Vancouver

Bibtex

@article{b2e73a50c3ce4afe8276eab94652d188,
title = "Modulating cardiac physiology in engineered heart tissue with the bidirectional optogenetic tool BiPOLES",
abstract = "Optogenetic actuators are rapidly advancing tools used to control physiology in excitable cells, such as neurons and cardiomyocytes. In neuroscience, these tools have been used to either excite or inhibit neuronal activity. Cell type-targeted actuators have allowed to study the function of distinct cell populations. Whereas the first described cation channelrhodopsins allowed to excite specific neuronal cell populations, anion channelrhodopsins were used to inhibit neuronal activity. To allow for simultaneous excitation and inhibition, opsin combinations with low spectral overlap were introduced. BiPOLES (Bidirectional Pair of Opsins for Light-induced Excitation and Silencing) is a bidirectional optogenetic tool consisting of the anion channel Guillardia theta anion-conducting channelrhodopsin 2 (GtACR2 with a blue excitation spectrum and the red-shifted cation channel Chrimson. Here, we studied the effects of BiPOLES activation in cardiomyocytes. For this, we knocked in BiPOLES into the adeno-associated virus integration site 1 (AAVS1) locus of human-induced pluripotent stem cells (hiPSC), subjected these to cardiac differentiation, and generated BiPOLES expressing engineered heart tissue (EHT) for physiological characterization. Continuous light application activating either GtACR2 or Chrimson resulted in cardiomyocyte depolarization and thus stopped EHT contractility. In contrast, short light pulses, with red as well as with blue light, triggered action potentials (AP) up to a rate of 240 bpm. In summary, we demonstrate that cation, as well as anion channelrhodopsins, can be used to activate stem cell-derived cardiomyocytes with pulsed photostimulation but also to silence cardiac contractility with prolonged photostimulation.",
keywords = "Humans, Optogenetics/methods, Channelrhodopsins/genetics, Myocytes, Cardiac/metabolism, Anions/metabolism, Cations",
author = "Barbora Schwarzov{\'a} and Tim St{\"u}demann and Muhammed S{\"o}nmez and Judith R{\"o}ssinger and Bangfen Pan and Thomas Eschenhagen and Justus Stenzig and Wiegert, {J Simon} and Torsten Christ and Florian Weinberger",
note = "{\textcopyright} 2023. The Author(s).",
year = "2023",
month = dec,
doi = "10.1007/s00424-023-02869-x",
language = "English",
volume = "475",
pages = "1463--1477",
journal = "PFLUG ARCH EUR J PHY",
issn = "0031-6768",
publisher = "Springer",
number = "12",

}

RIS

TY - JOUR

T1 - Modulating cardiac physiology in engineered heart tissue with the bidirectional optogenetic tool BiPOLES

AU - Schwarzová, Barbora

AU - Stüdemann, Tim

AU - Sönmez, Muhammed

AU - Rössinger, Judith

AU - Pan, Bangfen

AU - Eschenhagen, Thomas

AU - Stenzig, Justus

AU - Wiegert, J Simon

AU - Christ, Torsten

AU - Weinberger, Florian

N1 - © 2023. The Author(s).

PY - 2023/12

Y1 - 2023/12

N2 - Optogenetic actuators are rapidly advancing tools used to control physiology in excitable cells, such as neurons and cardiomyocytes. In neuroscience, these tools have been used to either excite or inhibit neuronal activity. Cell type-targeted actuators have allowed to study the function of distinct cell populations. Whereas the first described cation channelrhodopsins allowed to excite specific neuronal cell populations, anion channelrhodopsins were used to inhibit neuronal activity. To allow for simultaneous excitation and inhibition, opsin combinations with low spectral overlap were introduced. BiPOLES (Bidirectional Pair of Opsins for Light-induced Excitation and Silencing) is a bidirectional optogenetic tool consisting of the anion channel Guillardia theta anion-conducting channelrhodopsin 2 (GtACR2 with a blue excitation spectrum and the red-shifted cation channel Chrimson. Here, we studied the effects of BiPOLES activation in cardiomyocytes. For this, we knocked in BiPOLES into the adeno-associated virus integration site 1 (AAVS1) locus of human-induced pluripotent stem cells (hiPSC), subjected these to cardiac differentiation, and generated BiPOLES expressing engineered heart tissue (EHT) for physiological characterization. Continuous light application activating either GtACR2 or Chrimson resulted in cardiomyocyte depolarization and thus stopped EHT contractility. In contrast, short light pulses, with red as well as with blue light, triggered action potentials (AP) up to a rate of 240 bpm. In summary, we demonstrate that cation, as well as anion channelrhodopsins, can be used to activate stem cell-derived cardiomyocytes with pulsed photostimulation but also to silence cardiac contractility with prolonged photostimulation.

AB - Optogenetic actuators are rapidly advancing tools used to control physiology in excitable cells, such as neurons and cardiomyocytes. In neuroscience, these tools have been used to either excite or inhibit neuronal activity. Cell type-targeted actuators have allowed to study the function of distinct cell populations. Whereas the first described cation channelrhodopsins allowed to excite specific neuronal cell populations, anion channelrhodopsins were used to inhibit neuronal activity. To allow for simultaneous excitation and inhibition, opsin combinations with low spectral overlap were introduced. BiPOLES (Bidirectional Pair of Opsins for Light-induced Excitation and Silencing) is a bidirectional optogenetic tool consisting of the anion channel Guillardia theta anion-conducting channelrhodopsin 2 (GtACR2 with a blue excitation spectrum and the red-shifted cation channel Chrimson. Here, we studied the effects of BiPOLES activation in cardiomyocytes. For this, we knocked in BiPOLES into the adeno-associated virus integration site 1 (AAVS1) locus of human-induced pluripotent stem cells (hiPSC), subjected these to cardiac differentiation, and generated BiPOLES expressing engineered heart tissue (EHT) for physiological characterization. Continuous light application activating either GtACR2 or Chrimson resulted in cardiomyocyte depolarization and thus stopped EHT contractility. In contrast, short light pulses, with red as well as with blue light, triggered action potentials (AP) up to a rate of 240 bpm. In summary, we demonstrate that cation, as well as anion channelrhodopsins, can be used to activate stem cell-derived cardiomyocytes with pulsed photostimulation but also to silence cardiac contractility with prolonged photostimulation.

KW - Humans

KW - Optogenetics/methods

KW - Channelrhodopsins/genetics

KW - Myocytes, Cardiac/metabolism

KW - Anions/metabolism

KW - Cations

U2 - 10.1007/s00424-023-02869-x

DO - 10.1007/s00424-023-02869-x

M3 - SCORING: Journal article

C2 - 37863976

VL - 475

SP - 1463

EP - 1477

JO - PFLUG ARCH EUR J PHY

JF - PFLUG ARCH EUR J PHY

SN - 0031-6768

IS - 12

ER -