Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Standard

Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice. / Kim, Joon-Chul; Perez-Hernandez, Marta; Alvarado, Francisco J.; Maurya, Svetlana R.; Montnach, Jerome; Yin, Yandong; Zhang, Mingliang; Lin, Xianming; Vasquez, Carolina; Heguy, Adriana; Liang, Feng-Xia; Woo, Sun-Hee; Morley, Gregory E.; Rothenberg, Eli; Lundby, Alicia; Valdivia, Hector H.; Cerrone, Marina; Delmar, Mario.

I: Circulation, Bind 140, Nr. 12, 2019, s. 1015-1030.

Publikation: Bidrag til tidsskriftTidsskriftartikelForskningfagfællebedømt

Harvard

Kim, J-C, Perez-Hernandez, M, Alvarado, FJ, Maurya, SR, Montnach, J, Yin, Y, Zhang, M, Lin, X, Vasquez, C, Heguy, A, Liang, F-X, Woo, S-H, Morley, GE, Rothenberg, E, Lundby, A, Valdivia, HH, Cerrone, M & Delmar, M 2019, 'Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice', Circulation, bind 140, nr. 12, s. 1015-1030. https://doi.org/10.1161/CIRCULATIONAHA.119.039710

APA

Kim, J-C., Perez-Hernandez, M., Alvarado, F. J., Maurya, S. R., Montnach, J., Yin, Y., Zhang, M., Lin, X., Vasquez, C., Heguy, A., Liang, F-X., Woo, S-H., Morley, G. E., Rothenberg, E., Lundby, A., Valdivia, H. H., Cerrone, M., & Delmar, M. (2019). Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice. Circulation, 140(12), 1015-1030. https://doi.org/10.1161/CIRCULATIONAHA.119.039710

Vancouver

Kim J-C, Perez-Hernandez M, Alvarado FJ, Maurya SR, Montnach J, Yin Y o.a. Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice. Circulation. 2019;140(12):1015-1030. https://doi.org/10.1161/CIRCULATIONAHA.119.039710

Author

Kim, Joon-Chul ; Perez-Hernandez, Marta ; Alvarado, Francisco J. ; Maurya, Svetlana R. ; Montnach, Jerome ; Yin, Yandong ; Zhang, Mingliang ; Lin, Xianming ; Vasquez, Carolina ; Heguy, Adriana ; Liang, Feng-Xia ; Woo, Sun-Hee ; Morley, Gregory E. ; Rothenberg, Eli ; Lundby, Alicia ; Valdivia, Hector H. ; Cerrone, Marina ; Delmar, Mario. / Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice. I: Circulation. 2019 ; Bind 140, Nr. 12. s. 1015-1030.

Bibtex

@article{1969ea683dbe4699bd1e4fb3207af984,
title = "Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice",
abstract = "Background: Plakophilin-2 (PKP2) is classically defined as a desmosomal protein. Mutations in PKP2 associate with most cases of gene-positive arrhythmogenic right ventricular cardiomyopathy. A better understanding of PKP2 cardiac biology can help elucidate the mechanisms underlying arrhythmic and cardiomyopathic events consequent to PKP2 deficiency. Here, we sought to capture early molecular/cellular events that can act as nascent arrhythmic/cardiomyopathic substrates. Methods: We used multiple imaging, biochemical and high-resolution mass spectrometry methods to study functional/structural properties of cells/tissues derived from cardiomyocyte-specific, tamoxifen-activated, PKP2 knockout mice (PKP2cKO) 14 days post-tamoxifen injection, a time point preceding overt electrical or structural phenotypes. Myocytes from right or left ventricular free wall were studied separately. Results: Most properties of PKP2cKO left ventricular myocytes were not different from control; in contrast, PKP2cKO right ventricular (RV) myocytes showed increased amplitude and duration of Ca2+ transients, increased Ca2+ in the cytoplasm and sarcoplasmic reticulum, increased frequency of spontaneous Ca2+ release events (sparks) even at comparable sarcoplasmic reticulum load, and dynamic Ca2+ accumulation in mitochondria. We also observed early- and delayed-after transients in RV myocytes and heightened susceptibility to arrhythmias in Langendorff-perfused hearts. In addition, ryanodine receptor 2 in PKP2cKO-RV cells presented enhanced Ca2+ sensitivity and preferential phosphorylation in a domain known to modulate Ca2+ gating. RNAseq at 14 days post-tamoxifen showed no relevant difference in transcript abundance between RV and left ventricle, neither in control nor in PKP2cKO cells. Instead, we found an RV-predominant increase in membrane permeability that can permit Ca2+ entry into the cell. Connexin 43 ablation mitigated the membrane permeability increase, accumulation of cytoplasmic Ca2+, increased frequency of sparks and early stages of RV dysfunction. Connexin 43 hemichannel block with GAP19 normalized [Ca2+](i) homeostasis. Similarly, protein kinase C inhibition normalized spark frequency at comparable sarcoplasmic reticulum load levels. Conclusions: Loss of PKP2 creates an RV-predominant arrhythmogenic substrate (Ca2+ dysregulation) that precedes the cardiomyopathy; this is, at least in part, mediated by a Connexin 43-dependent membrane conduit and repressed by protein kinase C inhibitors. Given that asymmetric Ca2+ dysregulation precedes the cardiomyopathic stage, we speculate that abnormal Ca2+ handling in RV myocytes can be a trigger for gross structural changes observed at a later stage.",
keywords = "arrhythmogenic right ventricular cardiomyopathy, connexin43, plakophilin 2, right ventricle, sudden death",
author = "Joon-Chul Kim and Marta Perez-Hernandez and Alvarado, {Francisco J.} and Maurya, {Svetlana R.} and Jerome Montnach and Yandong Yin and Mingliang Zhang and Xianming Lin and Carolina Vasquez and Adriana Heguy and Feng-Xia Liang and Sun-Hee Woo and Morley, {Gregory E.} and Eli Rothenberg and Alicia Lundby and Valdivia, {Hector H.} and Marina Cerrone and Mario Delmar",
year = "2019",
doi = "10.1161/CIRCULATIONAHA.119.039710",
language = "English",
volume = "140",
pages = "1015--1030",
journal = "Circulation",
issn = "0009-7322",
publisher = "Lippincott Williams & Wilkins",
number = "12",

}

RIS

TY - JOUR

T1 - Disruption of Ca2+i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2-Deficient Mice

AU - Kim, Joon-Chul

AU - Perez-Hernandez, Marta

AU - Alvarado, Francisco J.

AU - Maurya, Svetlana R.

AU - Montnach, Jerome

AU - Yin, Yandong

AU - Zhang, Mingliang

AU - Lin, Xianming

AU - Vasquez, Carolina

AU - Heguy, Adriana

AU - Liang, Feng-Xia

AU - Woo, Sun-Hee

AU - Morley, Gregory E.

AU - Rothenberg, Eli

AU - Lundby, Alicia

AU - Valdivia, Hector H.

AU - Cerrone, Marina

AU - Delmar, Mario

PY - 2019

Y1 - 2019

N2 - Background: Plakophilin-2 (PKP2) is classically defined as a desmosomal protein. Mutations in PKP2 associate with most cases of gene-positive arrhythmogenic right ventricular cardiomyopathy. A better understanding of PKP2 cardiac biology can help elucidate the mechanisms underlying arrhythmic and cardiomyopathic events consequent to PKP2 deficiency. Here, we sought to capture early molecular/cellular events that can act as nascent arrhythmic/cardiomyopathic substrates. Methods: We used multiple imaging, biochemical and high-resolution mass spectrometry methods to study functional/structural properties of cells/tissues derived from cardiomyocyte-specific, tamoxifen-activated, PKP2 knockout mice (PKP2cKO) 14 days post-tamoxifen injection, a time point preceding overt electrical or structural phenotypes. Myocytes from right or left ventricular free wall were studied separately. Results: Most properties of PKP2cKO left ventricular myocytes were not different from control; in contrast, PKP2cKO right ventricular (RV) myocytes showed increased amplitude and duration of Ca2+ transients, increased Ca2+ in the cytoplasm and sarcoplasmic reticulum, increased frequency of spontaneous Ca2+ release events (sparks) even at comparable sarcoplasmic reticulum load, and dynamic Ca2+ accumulation in mitochondria. We also observed early- and delayed-after transients in RV myocytes and heightened susceptibility to arrhythmias in Langendorff-perfused hearts. In addition, ryanodine receptor 2 in PKP2cKO-RV cells presented enhanced Ca2+ sensitivity and preferential phosphorylation in a domain known to modulate Ca2+ gating. RNAseq at 14 days post-tamoxifen showed no relevant difference in transcript abundance between RV and left ventricle, neither in control nor in PKP2cKO cells. Instead, we found an RV-predominant increase in membrane permeability that can permit Ca2+ entry into the cell. Connexin 43 ablation mitigated the membrane permeability increase, accumulation of cytoplasmic Ca2+, increased frequency of sparks and early stages of RV dysfunction. Connexin 43 hemichannel block with GAP19 normalized [Ca2+](i) homeostasis. Similarly, protein kinase C inhibition normalized spark frequency at comparable sarcoplasmic reticulum load levels. Conclusions: Loss of PKP2 creates an RV-predominant arrhythmogenic substrate (Ca2+ dysregulation) that precedes the cardiomyopathy; this is, at least in part, mediated by a Connexin 43-dependent membrane conduit and repressed by protein kinase C inhibitors. Given that asymmetric Ca2+ dysregulation precedes the cardiomyopathic stage, we speculate that abnormal Ca2+ handling in RV myocytes can be a trigger for gross structural changes observed at a later stage.

AB - Background: Plakophilin-2 (PKP2) is classically defined as a desmosomal protein. Mutations in PKP2 associate with most cases of gene-positive arrhythmogenic right ventricular cardiomyopathy. A better understanding of PKP2 cardiac biology can help elucidate the mechanisms underlying arrhythmic and cardiomyopathic events consequent to PKP2 deficiency. Here, we sought to capture early molecular/cellular events that can act as nascent arrhythmic/cardiomyopathic substrates. Methods: We used multiple imaging, biochemical and high-resolution mass spectrometry methods to study functional/structural properties of cells/tissues derived from cardiomyocyte-specific, tamoxifen-activated, PKP2 knockout mice (PKP2cKO) 14 days post-tamoxifen injection, a time point preceding overt electrical or structural phenotypes. Myocytes from right or left ventricular free wall were studied separately. Results: Most properties of PKP2cKO left ventricular myocytes were not different from control; in contrast, PKP2cKO right ventricular (RV) myocytes showed increased amplitude and duration of Ca2+ transients, increased Ca2+ in the cytoplasm and sarcoplasmic reticulum, increased frequency of spontaneous Ca2+ release events (sparks) even at comparable sarcoplasmic reticulum load, and dynamic Ca2+ accumulation in mitochondria. We also observed early- and delayed-after transients in RV myocytes and heightened susceptibility to arrhythmias in Langendorff-perfused hearts. In addition, ryanodine receptor 2 in PKP2cKO-RV cells presented enhanced Ca2+ sensitivity and preferential phosphorylation in a domain known to modulate Ca2+ gating. RNAseq at 14 days post-tamoxifen showed no relevant difference in transcript abundance between RV and left ventricle, neither in control nor in PKP2cKO cells. Instead, we found an RV-predominant increase in membrane permeability that can permit Ca2+ entry into the cell. Connexin 43 ablation mitigated the membrane permeability increase, accumulation of cytoplasmic Ca2+, increased frequency of sparks and early stages of RV dysfunction. Connexin 43 hemichannel block with GAP19 normalized [Ca2+](i) homeostasis. Similarly, protein kinase C inhibition normalized spark frequency at comparable sarcoplasmic reticulum load levels. Conclusions: Loss of PKP2 creates an RV-predominant arrhythmogenic substrate (Ca2+ dysregulation) that precedes the cardiomyopathy; this is, at least in part, mediated by a Connexin 43-dependent membrane conduit and repressed by protein kinase C inhibitors. Given that asymmetric Ca2+ dysregulation precedes the cardiomyopathic stage, we speculate that abnormal Ca2+ handling in RV myocytes can be a trigger for gross structural changes observed at a later stage.

KW - arrhythmogenic right ventricular cardiomyopathy

KW - connexin43

KW - plakophilin 2

KW - right ventricle

KW - sudden death

U2 - 10.1161/CIRCULATIONAHA.119.039710

DO - 10.1161/CIRCULATIONAHA.119.039710

M3 - Journal article

C2 - 31315456

VL - 140

SP - 1015

EP - 1030

JO - Circulation

JF - Circulation

SN - 0009-7322

IS - 12

ER -

ID: 228368319