Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium

Standard

Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium. / Trautsch, Irina; Heta, Eriona; Soong, Poh Loong; Levent, Elif; Nikolaev, Viacheslav O; Bogeski, Ivan; Katschinski, Dörthe M; Mayr, Manuel; Zimmermann, Wolfram-Hubertus.

In: FRONT PHYSIOL, Vol. 10, 2019, p. 272.

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

Harvard

Trautsch, I, Heta, E, Soong, PL, Levent, E, Nikolaev, VO, Bogeski, I, Katschinski, DM, Mayr, M & Zimmermann, W-H 2019, 'Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium', FRONT PHYSIOL, vol. 10, pp. 272. https://doi.org/10.3389/fphys.2019.00272

APA

Trautsch, I., Heta, E., Soong, P. L., Levent, E., Nikolaev, V. O., Bogeski, I., Katschinski, D. M., Mayr, M., & Zimmermann, W-H. (2019). Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium. FRONT PHYSIOL, 10, 272. https://doi.org/10.3389/fphys.2019.00272

Vancouver

Bibtex

@article{b74f2bbefaff46c8b24dc4f89a89a398,
title = "Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium",
abstract = "Redox signaling affects all aspects of cardiac function and homeostasis. With the development of genetically encoded fluorescent redox sensors, novel tools for the optogenetic investigation of redox signaling have emerged. Here, we sought to develop a human heart muscle model for in-tissue imaging of redox alterations. For this, we made use of (1) the genetically-encoded Grx1-roGFP2 sensor, which reports changes in cellular glutathione redox status (GSH/GSSG), (2) human embryonic stem cells (HES2), and (3) the engineered heart muscle (EHM) technology. We first generated HES2 lines expressing Grx1-roGFP2 in cytosol or mitochondria compartments by TALEN-guided genomic integration. Grx1-roGFP2 sensor localization and function was verified by fluorescence imaging. Grx1-roGFP2 HES2 were then subjected to directed differentiation to obtain high purity cardiomyocyte populations. Despite being able to report glutathione redox potential from cytosol and mitochondria, we observed dysfunctional sarcomerogenesis in Grx1-roGFP2 expressing cardiomyocytes. Conversely, lentiviral transduction of Grx1-roGFP2 in already differentiated HES2-cardiomyocytes and human foreskin fibroblast was possible, without compromising cell function as determined in EHM from defined Grx1-roGFP2-expressing cardiomyocyte and fibroblast populations. Finally, cell-type specific GSH/GSSG imaging was demonstrated in EHM. Collectively, our observations suggests a crucial role for redox signaling in cardiomyocyte differentiation and provide a solution as to how this apparent limitation can be overcome to enable cell-type specific GSH/GSSG imaging in a human heart muscle context.",
author = "Irina Trautsch and Eriona Heta and Soong, {Poh Loong} and Elif Levent and Nikolaev, {Viacheslav O} and Ivan Bogeski and Katschinski, {D{\"o}rthe M} and Manuel Mayr and Wolfram-Hubertus Zimmermann",
year = "2019",
doi = "10.3389/fphys.2019.00272",
language = "English",
volume = "10",
pages = "272",
journal = "FRONT PHYSIOL",
issn = "1664-042X",
publisher = "Frontiers Research Foundation",

}

RIS

TY - JOUR

T1 - Optogenetic Monitoring of the Glutathione Redox State in Engineered Human Myocardium

AU - Trautsch, Irina

AU - Heta, Eriona

AU - Soong, Poh Loong

AU - Levent, Elif

AU - Nikolaev, Viacheslav O

AU - Bogeski, Ivan

AU - Katschinski, Dörthe M

AU - Mayr, Manuel

AU - Zimmermann, Wolfram-Hubertus

PY - 2019

Y1 - 2019

N2 - Redox signaling affects all aspects of cardiac function and homeostasis. With the development of genetically encoded fluorescent redox sensors, novel tools for the optogenetic investigation of redox signaling have emerged. Here, we sought to develop a human heart muscle model for in-tissue imaging of redox alterations. For this, we made use of (1) the genetically-encoded Grx1-roGFP2 sensor, which reports changes in cellular glutathione redox status (GSH/GSSG), (2) human embryonic stem cells (HES2), and (3) the engineered heart muscle (EHM) technology. We first generated HES2 lines expressing Grx1-roGFP2 in cytosol or mitochondria compartments by TALEN-guided genomic integration. Grx1-roGFP2 sensor localization and function was verified by fluorescence imaging. Grx1-roGFP2 HES2 were then subjected to directed differentiation to obtain high purity cardiomyocyte populations. Despite being able to report glutathione redox potential from cytosol and mitochondria, we observed dysfunctional sarcomerogenesis in Grx1-roGFP2 expressing cardiomyocytes. Conversely, lentiviral transduction of Grx1-roGFP2 in already differentiated HES2-cardiomyocytes and human foreskin fibroblast was possible, without compromising cell function as determined in EHM from defined Grx1-roGFP2-expressing cardiomyocyte and fibroblast populations. Finally, cell-type specific GSH/GSSG imaging was demonstrated in EHM. Collectively, our observations suggests a crucial role for redox signaling in cardiomyocyte differentiation and provide a solution as to how this apparent limitation can be overcome to enable cell-type specific GSH/GSSG imaging in a human heart muscle context.

AB - Redox signaling affects all aspects of cardiac function and homeostasis. With the development of genetically encoded fluorescent redox sensors, novel tools for the optogenetic investigation of redox signaling have emerged. Here, we sought to develop a human heart muscle model for in-tissue imaging of redox alterations. For this, we made use of (1) the genetically-encoded Grx1-roGFP2 sensor, which reports changes in cellular glutathione redox status (GSH/GSSG), (2) human embryonic stem cells (HES2), and (3) the engineered heart muscle (EHM) technology. We first generated HES2 lines expressing Grx1-roGFP2 in cytosol or mitochondria compartments by TALEN-guided genomic integration. Grx1-roGFP2 sensor localization and function was verified by fluorescence imaging. Grx1-roGFP2 HES2 were then subjected to directed differentiation to obtain high purity cardiomyocyte populations. Despite being able to report glutathione redox potential from cytosol and mitochondria, we observed dysfunctional sarcomerogenesis in Grx1-roGFP2 expressing cardiomyocytes. Conversely, lentiviral transduction of Grx1-roGFP2 in already differentiated HES2-cardiomyocytes and human foreskin fibroblast was possible, without compromising cell function as determined in EHM from defined Grx1-roGFP2-expressing cardiomyocyte and fibroblast populations. Finally, cell-type specific GSH/GSSG imaging was demonstrated in EHM. Collectively, our observations suggests a crucial role for redox signaling in cardiomyocyte differentiation and provide a solution as to how this apparent limitation can be overcome to enable cell-type specific GSH/GSSG imaging in a human heart muscle context.

U2 - 10.3389/fphys.2019.00272

DO - 10.3389/fphys.2019.00272

M3 - SCORING: Journal article

C2 - 31024328

VL - 10

SP - 272

JO - FRONT PHYSIOL

JF - FRONT PHYSIOL

SN - 1664-042X

ER -